Abstract
The extracellular nucleoside adenosine reduces tissue inflammation and is generated by irreversible dephosphorylation of AMP mediated by the ectonucleotidase CD73. The pro-inflammatory nucleotides ATP, NAD+ and cGAMP, which are produced in the tumor microenvironment (TME) during therapy-induced immunogenic cell death and activation of innate immune signaling, can be converted into AMP by ectonucleotidases CD39, CD38 and CD203a/ENPP1. Thus, ectonucleotidases shape the TME by converting immune-activating signals into an immune-suppressive one. Ectonucleotidases also hinder the ability of therapies including radiation therapy, which enhance the release of pro-inflammatory nucleotides in the extracellular milieu, to induce immune-mediated tumor rejection. Here we review the immune suppressive effects of adenosine and the role of different ectonucleotidases in modulating anti-tumor immune responses. We discuss emerging opportunities to target adenosine generation and/or its ability to signal via adenosine receptors expressed by immune and cancer cells in the context of combination immunotherapy and radiotherapy.
One sentence summary:
Adenosine generation as a rheostat of DNA damage-induced immune signaling in the tumor microenvironment
Introduction
In the last decade there has been unprecedented progress in leveraging the power of the immune system to treat cancer in patients. For the most part, this progress was the result of the identification of immune checkpoint receptors that inhibit the function of T cells and are co-opted to protect tumors from immune-mediated rejection (1). For the majority of cancer patients, however, blocking these immune checkpoint receptors or their ligands is not sufficient. Multiple strategies aimed at promoting an inflamed tumor mircoenvironment to increase responses to immune checkpoint inhibitors (ICI) have been tested. These include the use of radiotherapy, conventional or targeted chemotherapy, and biological agents that mimic pathogen-derived signals such as Toll-like Receptor (TLR) agonists and oncolytic agents (2). Overall, the success of these combinations has been modest, suggesting that it is essential to identify and target immunosuppressive barriers, some of which can be exacerbated by pro-inflammatory treatments.
A large body of preclinical evidence supports the ability of radiotherapy to increase responses to ICI (3). Mechanistically, radiation induces an immunogenic cell death associated with release of pro-inflammatory signals such as adenosine triphosphate (ATP) (4, 5), and activates canonical pathways of viral defence, mediated by exposure of nuclear and mitochondrial DNA to the cytosolic sensor cyclic GMP-AMP synthase (cGAS). cGAS catalyzes the production of the cyclic dinucleotide cyclic GMP-AMP (cGAMP), which binds to stimulator of IFN genes (STING) leading to the downstream activation of type I interferon (IFN-I) and NF-ΚB pathways in the cancer cells and in innate immune cells in the tumor microenvironment (TME) (6). Activation of IFN-I in dendritic cells (DCs) in the TME of the irradiated tumor has been shown to occur via the uptake of tumor DNA as well as cGAMP, which is exported from irradiated cancer cells to the extracellular compartment (7, 8). Overall, extracellular ATP and cGAS-STING pathways promote cross-presentation of tumor antigens by DCs to T cells with priming and expansion of tumor-specific T cells, a process that is facilitated and sustained by ICI (9).
In the clinic, the ability of radiotherapy to induce/enhance responses to ICI is supported by results of early phase trials (10–13). However, the initial assumption that radiotherapy could be broadly beneficial in enhancing ICI responses has been proven incorrect (14). Although there remain multiple open questions pertaining to the dose and sequencing of radiation with ICI and to the optimal radiation target (15), recent preclinical data highlight the importance of ectonucleotidases expressed in the TME and adenosine signaling as a potential immunosuppressive mechanism that needs to be countered to improve the therapeutic efficacy of combinations of radiotherapy and ICI (16, 17). In this review we provide an overview of ectonucleotidases involved in the degradation of extracellular ATP into adenosine (ADO) and regulation of the cGAS-STING pathway in the TME (Figure 1). In addition, we discuss emerging opportunities to modulate this balance to improve responses to combinations of radiotherapy and other pro-inflammatory agents with ICI.
Figure 1. The immunogenicity of radiotherapy is regulated by ectonucleotidases.
1. Radiotherapy -induced cytosolic DNA leads to activation of cGAS that generates cGAMP, part of which can be exported into the extracellular compartment where it activates STING in immune cells. Downstream IFN-I promotes cross-presenting dendritic cell (DC) activation and priming of anti-tumour T cells. 2. ENPP1 converts cGAMP into AMP, generating a substrate for adenosine generation by CD73. 3. ATP released by irradiated cancer cells binds to P2X7R on DCs, promoting production of pro-inflammatory cytokines. 4. CD39 converts ATP into ADP and AMP which is then converted into adenosine by CD73. Adenosine exerts immune suppression by binding to adenosine receptors A2AR and A2BR on immune cells. 5. Extracellular NAD+ also contributes to adenosine generation by sequential conversion into ADPR mediated by CD38, into AMP mediated by ENPP1, and into adenosine via CD73. In addition, NAD+ serves as a substrate for ADP-ribosyltransferases (ARTs) including ART1 which mono-ADP-ribosylate (MARylate) the P2X7R on T cells to induce NAD-induced cell death (NICD). Importantly CD38 and CD39 expressed by immune cells can offset NICD and ATP-induced cell death by catabolizing NAD+ and ATP respectively, thus providing cytoprotective roles.
Overview of the adenosine pathway
Extracellular purines play a critical role in various processes such as cell death, phagocytosis, inflammasome activation, antigen presentation, and cell migration. Purines activate purinergic receptors, divided into three different families: P1 receptors (P1Rs), P2Y receptors (P2YRs), and P2X receptors (P2XRs). P1Rs are G-protein-coupled receptors (GPCRs) activated by adenosine and comprise four subtypes (A1, A2A, A2B, and A3), while P2YRs are GPCRs activated by di- and triphosphate-nucleotides, including ATP, and include at least eight subtypes. P2XRs are ATP-gated ion channels with seven subtypes. Compared with other P2X receptors, P2XR7 has a relatively low affinity to ATP, and its maximal activity is achieved at ATP concentrations of 100–1000 μM (18).
ADO is a potent immunosuppressive metabolite that carries its function essentially through the activation of A2A and A2B GPCRs expressed on immune cells (Figure 2). In solid tumors, ADO accumulates as a result of the hydrolysis of extracellular ATP by ectonucleotidases, reaching concentrations up to 100 μM (19, 20). Extracellular ATP is generally low in most tissue (10–100 nM) but rises up to 1,000-fold in response to tissue damage, hypoxia, ischemia or cell death (21). This accumulation of extracellular ATP is an important danger signal that helps initiate inflammatory processes mainly via the activation of P2Y and P2X receptors expressed on immune cells (Figure 2). In tumors, extracellular ATP plays a key role in promoting antigen presentation and adaptive anti-tumor responses (reviewed in (22)).
Figure 2. Immune regulation through extracellular adenosine (ADO) and adenosine triphosphate (ATP).
In the tumor microenvironment, ADO activates A2AR and A2BR to promote the production of IL-10 and VEGF by myeloid cells, thereby suppressing antigen presentation. In contrast, ATP activates P2X7R and P2Y2R to enhance production of proinflammatory IL-1β, IL-18, IL-6 and IL-12. In CD8+ effector T cells (CD8), A2AR inhibits IFN-γ, TNF-α and proliferation, while P2X7R and P2X4R promote T cell proliferation and immune memory. A2AR further enhances the suppressive function of T regulatory cells (Treg), while P2X7R inhibits FOXP3 expression. In CD4+ T effector cells (CD4), A2AR inhibits IFN-γ, TNF-α and proliferation, while activation of P2X7R promotes T follicular helper (Tfh) function. ADO inhibits B cells via A2AR, while P2X7R promotes B cell activation. Finally, activation of A2AR and/or P2X7R in natural killer (NK) cells inhibit their function.
Several mechanisms of ATP release have been described. These include pannexin channels (PANX), connexins (CONX), ATP-binding cassette transporters and P2X7 purinergic receptors (23, 24). Extracellular vesicles or microparticles are another characterized pathway for ATP release (25). ATP and adenosine diphosphate (ADP) can also be released by platelets through an exocytosis-mediated mechanism (26). Once in the extracellular environment, ATP can activate purinergic signaling via specialized ion-gated channels named P2X receptors, selective G-protein-coupled receptors named P2Y receptors, or can be hydrolyzed into ADO and its mono- or di-phosphorylated derivatives by specialized ectonucleotidases (27).
CD39 and CD73 constitute key ectonucleotidases which promote the metabolism of pro-inflammatory ATP into immunosuppressive ADO. CD39 (ecto-nucleoside triphosphate diphosphohydrolase-1; ENTPD1) hydrolyzes extracellular ATP/ADP into AMP (28), while CD73 converts AMP into extracellular ADO (29). Another important ectonucleotidase able to hydrolyse extracellular ATP is ecto-nucleotide pyrophosphate phosphodiesterase-1 (ENPP1, also known as CD203a, PC-1) (30). Additional ADO-generating pathways involving CD38 and ENNP1 are discussed below.
Pro-inflammatory functions of extracellular ATP
ATP and immunogenic cell death
In the TME, dying cells release ATP through caspase- and PANX1-dependent lysosomal exocytosis to alarm the immune system (31). ATP release is an essential component of immunogenic cell death (ICD), which refers to regulated mechanisms of cell death able to stimulate adaptive immune responses (32). Importantly, various cytotoxic agents as well as radiotherapy can cause ICD and enhance adaptive anti-tumor immunity (reviewed in (33)).
During ICD, extracellular ATP stimulates the recruitment of DCs and macrophages via P2Y2 receptors and cooperates with TLR signaling to activate the NLRP3 inflammasome via P2X7 receptors. Upon ATP-P2X7R and TLR signaling, NLRP3 associates with ASC adaptor protein to recruit and activate caspase-1, ensuing cleavage of pro-IL-1β and pro-IL-18 to their secreted forms. Amongst its pleiotropic effects, IL-1β enhances T cell granzyme B expression and proliferation, while IL-18 stimulates innate lymphocytes and antigen-experienced T cells (34). In support of a critical role for extracellular ATP in anti-tumor immunity, targeted blockade of CD39 significantly enhances the effect of ICI in mice in a mechanism dependent on P2X7R, ASC, NLRP3 and IL-1/IL-18 (35).
ATP and macrophages
Macrophages sense tissue damage through purinergic receptors and migrate toward sources of extracellular ATP via P2Y2 receptors (36). Following stimulation with ATP, macrophages further upregulate phosphatidylserine and release several cytokines, including IL-6, IL-12 and CXCL5 (Figure 2) (37–39). ATP signaling via P2X7R eventually upregulates CD39 expression on macrophages, thereby downregulating pro-inflammatory effects (38). Blocking CD39 on macrophages thus constitutes an effective means to enhance IL-12 and tumour-necrosis factor-α (TNF-α) release, while decreasing IL-10 production (40). P2X7R signaling may also promote gasdermin-D (GSDMD)-dependent pyroptosis of macrophages (41), which has been suggested to promote anti-tumor immunity (42).
ATP and T cells
ATP has different effects on T cells. Upon T cell receptor (TCR) signaling, ATP is rapidly released from activated T cells through PANX1 channels. This release of ATP serves T cells in an autocrine and paracrine manner to amplify intracellular Ca2+ signaling by activating P2X4 and P2X7 receptors. Extracellular ATP thus stimulates human T cell proliferation. Notably, blocking P2X4R significantly prevents T cell proliferation and migration (43). In vitro work further suggests that basal ATP release from T cells and P2X4R signaling prime unstimulated T cells and subsequently impact their activation upon antigen encounter (44).
Extracellular ATP also impacts T cell-mediated immunological memory and antigen presentation by activating P2X7R (45). P2X7R signaling in T cells is required for the establishment, maintenance and functionality of long-lived central and tissue-resident memory T cells. Using adoptive transfer studies in LCMV-infected mice of wild type versus P2rx7−/− TCR-transgenic CD8+ T cells (P14 cells), Borges da Silva et al. observed that P2X7R deficiency was associated with a significant loss of CD62L+ central memory T cells (TCM) (46). In the early phase of memory T cell differentiation, P2rx7−/− P14 cells showed a selective decline in memory precursor effector cells (MPECs). Mechanistically, loss of P2X7R signaling in MPECs was associated with significantly reduced mitochondrial respiration and aerobic glycolysis, translating to lower T cell survival and reduced recall expansion in vivo.
P2X7R signaling also regulates the generation and maintenance of tissue-resident memory T cells (TRM), which represent specialized memory T cells that do not recirculate. In a mouse model of LCMV infection, P2X7R was found to be required to support TRM generation of adoptively transferred antigen-specific T cells. Mechanistically, P2X7R was shown to enhance expression of TGF-β receptor and CD103 on TRM cells (47). On the other hand, P2X7R expression may limit the accumulation and function of tumor-infiltrating T cells (TILs). Romagnani et al. notably demonstrated that adoptively transfered tumor-specific T cells with deletion of P2rx7 are more effective at reducing tumor growth in mice (48). Mechanistically, activation of P2X7R induced senescence of the TILs.
Extracellular ATP not only promotes antigen presentation and T cell function, it also suppresses T regulatory cells (Tregs), which express higher levels of P2X7R than conventional T cells. P2X7R signaling in Tregs has been shown to trigger a decrease of FOXP3 protein levels and to render Tregs more susceptible to Th17 conversion (49).
P2X7R signaling also plays an important role in T follicular helper (Tfh) cell responses. During immunization, P2X7R signaling promotes expansion of Tfh cells, antigen-specific CD4+ T cells and germinal centers. In the context of immunopathogenic responses, however, where extracellular ATP levels are presumably higher, P2X7R signaling in Tfh has been proposed to limit Tfh survival via GSDMD-mediated pyroptosis (50). In support of a role for ATP signaling in promoting Tfh, ectopic expression of BCL6, the central regulator of Tfh differentiation, represses CD39 (ENTPD1) transcription. Consistent with these observations, CD39 overexpression in CD4+ T cells decreased Tfh development in vitro. Together, these studies suggest that targeting the CD39-CD73 axis may provide a means to enhance antibody responses, for example following vaccination (51).
ATP and NK cells
Intriguingly, evidence suggest that P2X7R activation on NK cells restricts their anti-tumor activity. In vitro, activation of P2X7R with ATP decreased human NK cell cytotoxicity against K562 target cells (52). Injection of B16 melanoma cells in CD8-depleted WT and P2×7r−/− mice also revealed a role for P2X7R in suppressing NK cell-mediated immune surveillance (53).
ATP and B cells
Activation of the P2X7 receptor by extracellular ATP can modulate B cell receptor signaling and influence B cell activation, proliferation, and differentiation. P2X7R stimulation has been shown to promote B cell activation (54), and B cell expression of CD39 is increased on activation in the presence of ATP(55). The ability of B cells to hydrolyze ATP also contribute to immunosuppression. Tumor-infiltrating B cells indeed contribute to increase immunosuppressive ADO levels (56). Septic B cells have also been shown to suppress macrophage-mediated bacterial killing via A2A receptors (57).
Immunosuppression via extracellular ADO
While extracellular ATP can stimulate both innate and adaptive immune responses, tumors are proficient at hydrolysing ATP into ADO. Accordingly, ectonucleotidases are often upregulated in the TME as a result of tissue hypoxia (58), inflammatory cytokines (e.g. TNF-α) (59) and tissue repair pathways (e.g. TGF-β and EMT) (60). Activation of oncogenic pathways, for instance mediated by mutations in KRAS, BRAF, EGFR or ALK-rearrangement, can furthermore upregulate the expression of ectonucleotidases by tumor cells (61). Additionally, radiotherapy has been shown to increase the expression by cancer cells of CD73 and other ectonucleotidases (16, 17).
ADO receptor signaling
Extracellular ADO exerts its effects thought four specific GPCRs. A1 (ADORA1), A2A (ADORA2A) and A2B (ADORA2B) ADO receptors are highly conserved across mammalian species, while A3 (ADORA3) is more variable (reviewed in (62)). In humans, A1, A2A and A3 receptors bind ADO with high affinity, while A2BR has a significantly lower affinity (Kd of 15 μM) that restricts its activation to pathological conditions when ADO concentration is high, such as may occur in the irradiated TME (16). A2AR and A2BR are generally Gs-coupled and therefore trigger intracellular cAMP accumulation, activating protein kinase A (PKA) and EPAC pathways. In contrast, A1 and A3 receptors are preferentially coupled to Gi/o proteins, inhibiting intracellular cyclic AMP production. A2BR and A3R can furthermore recruit Gq/11 proteins and trigger the phospholipase C (PLC) pathways, while all four receptors can induce mitogen-activated protein kinase (MAPK) and JNK pathways.
Landmark studies from Sitkovsky and colleagues established the critical importance of A2AR signaling for downregulation of systemic inflammation and associated tissue damage. Using experimental models of liver damage and sepsis, Otha et al. demonstrated that global A2AR deficiency in mice was associated with a substantial increase in systemic TNF-α, IFN-γ and IL-12 inflammatory cytokines upon endotoxin injection (63).
Interestingly, inosine produced from extracellular ADO by adenosine deaminase can also activate A2A receptors. In contrast to ADO, however, inosine induces a biased activation of the receptor towards increased ERK1/2 signalling over PKA signaling (64). This may account for the distinct impact of ADO and inosine on tumor immunity, although this has not been strictly investigated. Intriguingly, inosine production by the gut microbiome has been shown to stimulate anti-tumor T cells via A2A receptors when sufficient co-stimulation in provided (65).
ADO and myeloid cells
Myeloid-specific conditional deletion studies have revealed the importance of A2A and A2B receptors in the regulation of innate immunity (66). ADO signaling promotes the expression of tolerogenic cytokines by myeloid cells and suppresses the antigen-presenting function of macrophages and DCs. In murine tumors, targeting A2AR with a small molecule inhibitor, AZD4635, significantly enhances the expression of genes associated with antigen processing (Cd1d2, H2-Ea-ps, Cd207), macrophage and DC function (Fcer1a, Cr2), and upregulates the expression of antigen-presenting MHC class II and costimulatory CD86 molecules on macrophages and DCs (67). Further highlighting the impact of A2AR signaling on antigen presentation, in vitro co-culture assays revealed that human DCs exposed to the adenosine analog 5′-N-ethylcarboxamide adenosine (NECA) are significantly impaired in their ability to cross-prime tumor-specific human T cells (>90% reduction), an effect that is completely reversed by AZD4635.
Activation of A2BR also favors tolerogenic DCs and macrophages. Early work in mouse monocyte-derived DCs revealed that A2BR signaling inhibits TNFα, IL-12 and CD86 expression while increasing IL-10 production (68). More recently, using conditional deletion of A2B in LysM+ or CD11c+ cells, Cekic and colleagues demonstrated that A2BR activation in tumor-infiltrating myeloid cells suppresses CD86 and MHC class II expression, and was associated with a significant increase in B16 tumor growth. Notably, ex vivo assays revealed that A2BR-deficient DCs were significantly more effective in driving antigen-specific T-cell responses (66).
ADO and T cells
The A2AR is significantly upregulated upon T cell activation, rendering T cells susceptible to ADO-mediated immunosuppression. Activation of A2AR suppresses TCR, CD28 and IL-2 signalling, thus inhibiting T cell proliferation, survival and cytokine production. Granzyme-mediated cytotoxic functions, however, appear less affected by A2AR signaling, as demonstrated in CRISPR/Cas9 knockout studies (69). Targeting A2A receptors with selective antagonists notably triggers compensatory upregulation of other immune checkpoints, including PD-1, TIGIT, LAG-3 and TIM-3, thus providing a rationale for combination therapies (70). Recent studies of chimeric antigen receptor (CAR) T cells suggest that A2AR distinctively regulates CD8+ versus CD4+ T cells, and that TNF-α production remains partially suppressed by NECA in A2AR-deficient CAR T cells, suggesting a possible role for A2B receptor in ADO-induced T cell inhibition (69).
In humans, CD73 is expressed on approximately 40–80% of circulating naïve CD8+ T cells and 20–40% of circulating naïve CD4+ T cells. Upon activation, CD73 is downregulated and/or released from the cell surface (71). However, it is re-expressed on a smaller fraction of central and effector memory T cells (72). CD39 expression, on the other hand, is induced in T cells by TCR signaling and RUNX3 transcriptional activity (73). CD39 is further upregulated in exhausted T cells. CD39high PD-1high tumor-infiltrating T cells are notably enriched for tumor antigen specificity (74). Chronic LCMV responses in mice suggest a role for CD39-mediated purinergic signaling in the regulation of T cell exhaustion (75). Of interest, a recent report revealed a unique role for IL-4 in downregulating CD39 expression on activated T and B cells (73).
Recently, it has been shown that upon activation, human CD8+ T cells release CD73-containing extracellular vesicles (EVs) that generate ADO as a regulatory mechanism (71). Since inhibition of phospholipase or metalloproteinases failed to prevent the loss of membrane-bound CD73 on activated T cells, EV release appears to be the mechanism behind CD73 downregulation from the cell surface in response to TCR activation. Although it remains unclear whether CD39 is also present in T cell-derived EVs, prior studies reported the release of CD39- or CD73-containing EVs from B cells, Tregs and mesenchymal stromal cells (MSCs) (76, 77). Taken together, this suggests a previously underappreciated role for EV-dependent ADO production in the control of immune responses.
CD73 expression is enriched in antigen-experienced memory T cells (TMEM), which constitute a functionally distinct subset that declines with age (72). CD73+ TMEM notably show increased ability to differentiate into TRM, as demonstrated using adoptive transfer studies during LCMV infection in mice (72). Interestingly, an increased expression of ribosomal genes was also observed in CD73+ TMEM, a characteristic associated with effector rather than memory T cells. Although it remains to be investigated whether the increased functionality and TRM differentiation potential of CD73+ TMEM is related to ADO signaling, CD73 appears to identify circulating TMEM prone to differentiate into TRM cells.
In Tregs, ADO signaling through the A2AR promotes proliferation, production of immunosuppressive TGF-β and IL-10, and expression of immune-checkpoint receptors, overall enhancing their immunoregulatory activity in vitro (78). A2AR signaling in vivo has been involved in the emergence of induced Tregs during autoimmune disease (79), and exposing Tregs to an A2AR agonist has been shown to enhance the prevention of ischemia-reperfusion injury in mice following adoptive cell transfer (80).
Tregs can also participate in ATP hydrolysis, although CD39 and CD73 are not uniformly expressed, and rarely co-expressed, on human Tregs. There is no correlation between the expression levels of FOXP3, HELIOS, CD39 and CD73 in peripheral human Tregs (81, 82). During in vitro polarization of naïve T cells into induced Tregs (iTregs), Gerner et al. (81) recently demonstrated that mTOR signaling and TGF-β/SOX4 signaling were required for CD39 expression on iTregs. CD73, on the other hand, is generally absent from human Tregs, but can be induced in response to IL-2. Accordingly, Tregs from melanoma patients receiving high-dose IL-2 co-express CD39 and CD73 (83). Similarly, flow-sorted CD4+ CD25hiCD127lo human Tregs stimulated with anti-CD3/CD28 and expanded in the presence of high-dose IL-2 (500 U/mL) co-express CD39 and CD73 and display superior suppressive function when compared to matched ex vivo Tregs that do not express CD73 (82).
ADO and NK cells
In mice conditionally deficient for A2AR in NK cells, the maturation of NK cells is enhanced and terminally mature NK cells display increased effector function (84). In the absence of A2AR signaling, NK cells show reduced expression of IL18R1 and KIT genes, which may in part explain better tumor control (85). Tumor-infiltrating NK cells also upregulate CD73 and this is associated with acquired expression of IL-10 and TGF-β production via STAT3 signaling (86). Ex vivo, NK cells can acquire CD73 expression upon coculture with tumor cells in a contact-dependent manner, in a mechanism that may involve 41BB activation on NK cells, although alternative pathways may also be involved.
ADO and B cells
ADO signaling inhibits human B cell proliferation in vitro when stimulated by CD40L and IL-4 (87). The anti-proliferative effect of ADO on B cells is partly reversed by A3 receptor blockade, but appears independent of A2AR. ADO also inhibits human B cells activation with anti-μ-F(ab’)2, evidenced by reduced phosphorylation of Bruton’s tyrosine kinase (BTK) (56). Interestingly, BTK signaling upregulates CD39 expression on B cells.
Targeting CD73 with the blocking mAb ciforadenant has been reported to increase B cell activation, evidenced by upregulation of CD69, CD83, CD86 and MHC II, and to induce differentiation into plasmablasts (88). Intriguingly, these effects were found to be independent of CD73 enzymatic activity or ADO signaling, suggesting transmission of an activation signal downstream of CD73 when ligated by antibodies, as initially proposed by L. Thompson and colleagues (89).
Alternative pathways of ADO generation
In addition to ATP, other pro-inflammatory nucleotides can be converted into extracellular ADO. This occurs independently of CD39, involving ectonucleotidases CD38 and ENPP1, but still requires CD73 for the final step of catabolizing AMP into ADO.
CD38 and its role in cancer
CD38 was first characterized in T and B lymphocytes, where its expression has been used as a marker of differentiation (90, 91), but it is also expressed by NK and myeloid cells. CD38 has several enzymatic properties, firstly as a cyclase converting NAD+ to cyclic adenosine diphosphate ribose (cADPR) and secondly as a glycohydrolase (NADase) converting NAD+ and cADPR to ADPR (92).
CD38 is the dominant NADase in mammals making it a major regulator of intracellular and extracellular NAD+ levels. Chini and colleagues, who were some of the first to determine the enzymatic properties of CD38, showed that NAD tissue levels are 10–20 fold higher in CD38-deficent compared with CD38-proficient mice (92). CD38-mediated intracellular regulation of NAD+ has broad implications for cellular energy transfer, as well as for non-redox reactions including NAD-dependent post translational protein modifications including mono- and poly-ADP-ribosylation, mediated by mono-ADP-ribosyltransferases (ARTs) and poly-ADP-ribose-transferases (PARPs), respectively (93–95). In 2013, Malavasi and coworkers demonstrated that ADPR generated by CD38 is hydrolysed by ENPP1 into AMP, which is further dephosporylated into adenosine by CD73 (96, 97).
CD38 is a driver of tumour progression in many hematopoietic cancers. It is highly expressed by multiple myeloma (MM) cells as well as by several myeloid and lymphocytic leukemia cells. In MM, CD38 antibody treatment has proven to be effective by directly killing tumour cells, as well as by eliminating myeloid-derived suppressor cells (MDSCs), Tregs and regulatory B cells (Bregs), resulting in reduced immune suppression (98). Moreover, in the hypoxic and acidic bone marrow niche, the CD38-mediated non-canonical adenosine pathway is believed to operate more effectively than the classical pathway providing an explanation for the anti-tumour effect of CD38-inhibition in this cancer (99).
The role of CD38 in progression of solid tumors remains incompletely elucidated. Several independent studies have shown an association between CD38 expression and anti-tumour immune responses, although it remains unclear whether CD38 expression promotes or counteracts immune cell activity, which may depend on which cells it is expressed. Analysis of CD38 expression in a TCGA human lung cancer patient cohort showed a positive correlation with cytolytic T cell score (100). Similarly, RNA sequencing and immune fluorescence analysis of biopsies from metastatic castration-resistant prostate cancer (mCRPC) patients revealed that mRNA expression of CD38 was associated with active immune gene signature but also with T cell exhaustion and adenosine signalling. In these samples, CD38 was primarily expressed on MDSCs and B cells whose infiltration was a poor prognostic factor for OS (101). However, an inverse correlation between CD38 expression and prostate cancer progression has also been observed and attributed to metabolic reprogramming of tumour cells due to reduced NAD+ pools resulting in reduced proliferation (102). Furthermore, in pre-clinical models of esophageal cancer, CD38+ MDSCs were shown to have higher T cell-suppressive capacity than CD38- MDSCs and promote tumour progression (103). Conversely, in studies of hepatocellular carcinoma (HCC), tumour-infiltrating macrophages were predominantly M1-polarized, secreted pro-inflammatory cytokines and their density correlated with improved prognosis following surgery (104). In line with these findings, in HCC patients treated with ICI the overall response rate was higher in patients with high tumour infiltration of CD38+CD68+ macrophages (105).
CD38 expression on immune and cancer cells is upregulated by soluble factors present in the tumour microenvironment including TNF-α, trans-retinoic acid as well as IFN-γ and IFN-β indicating a physiological response to inflammatory conditions (100, 106). Increased CD38 expression has also been observed in colorectal cancer patients following chemotherapy (107), and in melanoma patients following anti-PD-1 therapy (100). Further, CD38 expression on human and murine breast cancer cells is upregulated by exposure to ionizing radiation in a dose-dependent manner (16).
In preclinical lung cancer models, Chen and colleagues reported that CD38-mediated adenosine production promotes acquired tumour resistance to anti-PD-1/PD-L1 therapy (100). In their models, PD-1/PD-L1-mediated upregulation of CD38 on tumour cells was driven by trans-retinoic acid, and IFN-β and tumour knockdown or therapeutic blockade of CD38 synergized with PD-L1 inhibition to promote CD8 T cell-dependent anti-tumour immunity. They further show that CD38 expression correlated with adenosine production, which was reduced upon anti-CD38 antibody treatment. Translation of such combination of CD38 blocking antibodies (daratumumab and isatuximab) with PD-1/PD-L1 blockade in patients with solid tumours however failed to demonstrate a therapeutic benefit, and some studies were terminated early due to increased mortality in the combination arm compared with ICI alone (108–110). These results suggest the need for an improved understanding of the role of CD38 in the TME.
Through its cyclase function, CD38 generates cADPR, which serves as a second messenger for Ca2+ mobilization during T cell activation. Together with the finding that CD38 clusters at the immune synapse upon TCR engagement with antigen-presenting cells, this suggests that CD38 is a regulator of T cell function (111). CD38 has also been shown to promote DC trafficking to lymph nodes in response to chemokines, and in CD38-deficient mice T cell priming and T cell-dependent humoral responses are reduced (112). Thus, CD38 inhibition in the context of ICI could potentially interfere with the priming and/or effector phase of the anti-tumor immune response.
Furthermore, ARTs use extracellular NAD+ (eNAD) as a substrate to mono-ADP-ribosylate P2X7R expressed on T cells resulting in NAD-induced cell death (NICD). In preclinical studies this process has been shown to operate in tissues during inflammation to selectively eliminate TRM that are not antigen-activated, as well as Tregs (113–115). Krebs et al. showed that ART2-mediated mono-ADP-ribosylation of T cells following eNAD exposure was higher in CD38-deficient mice compared to wild type mice (116). While ART2 is not expressed in humans due to premature stop codons, ART1 expression by lung cancer cells has recently been characterized as an immune resistance mechanism in NSCLC (117, 118). In an NSCLC mouse model high expression of ART1 by the cancer cells was associated with reduced CD8 T cell infiltration and mice survival. In vitro, ART1+ cancer cells induced NICD of P2X7R+ CD8 T cells, which was exacerbated by CD38 blockade, indicating a protective role of CD38 expressed by P2X7R+ CD8 T cells against NICD. Consistently, P2X7R+ CD8 T cells infiltrating ART1-expressing human lung tumors were enriched for CD38 expression (118). Thus, in solid tumours, particularly following radiotherapy, where eNAD concentrations are high, the function of CD38 as an NADase confers a cytoprotective role for T cells to avoid NICD. These data suggest the possibility that in patients with ART1-expressing tumours CD38-inhibition in combination with ICI could have a detrimental effect by augmenting NICD of tumour-infiltrating CD8 T cells.
ENPP1 and its role in cancer
ENPP1 generates AMP through hydrolysis of ATP, ADPR or through direct hydrolysis of NAD+ into AMP which is subsequently converted into adenosine by CD73 (97). Recently, ENPP1 has been shown to suppress innate immune activation in the TME by hydrolysing extracellular cGAMP (119). cGAMP is synthesized by cGAS upon binding to DNA in the cytosolic compartment, where it is present during viral infection or due to genetic instability and the activation of the DNA damage response in neoplastic cells (120). cGAMP binds with high affinity to the endoplasmatic reticulum receptor STING, which subsequently triggers a signalling pathway resulting in transcription of IFN-I and other pro-inflammatory cytokines. Intracellular cGAMP levels are regulated by continuous cellular import and export mediated by transporters (121, 122), including the newly described ATP binding cassette subfamily C member 1 (ABCC1) transporter (123). Intercellular exchange of cGAMP via gap junctions has also been described (124). To date, the only identified hydrolase of cGAMP is ENPP1 which is an extracellular membrane-bound enzyme. By reducing the ability of chromosomally unstable and DNA-damaged cells to propagate immunogenic signalling via cGAMP, while, through this process producing immunosuppressive adenosine-precursors in the form of AMP, ENPP1 is being increasingly recognized as an immune checkpoint and a potential immunotherapeutic target.
High ENPP1 expression in breast cancer has been associated with poor prognosis (125) and increased bone metastasis (126). Carozza and colleagues recently showed that extracellular cGAMP export from irradiated breast tumours dictates radiation-induced immunogenicity in a STING-dependent manner, in large part through activation of conventional type I DC (cDC1). In the aggressive 4T1 triple negative murine mammary cancer model, genetic deletion of ENPP1 in implanted 4T1 tumours, or administration of a cell impermeable ENPP1 inhibitor, resulted in reduced tumor progression (8). Interestingly, radiation increases surface expression of ENPP1 on 4T1 cells, suggesting that activation of cGAS by radiation is coupled with the induction of a counter-regulatory mechanism aimed at reducing immune activation (16).
Loss of tumour-intrinsic ENPP1 by genetic knockout in 4T1 and other tumors was shown to reduce adenosine production in vitro while in vivo it was associated with increased tumor infiltration of activated T cells and increased response to ICI (119). The anti-tumour or pro-tumour effect of ENNP1 deletion or overexpression, respectively, were reduced in STING-deficient mice indicating that ENNP1 promotes tumour progression primarily by preventing cGAMP activation of STING in host cells. Further, significantly higher ENNP1 cancer cell expression was measured in distant metastasis compared with primary tumours. Interestingly, the opposite expression pattern was observed with CD39 potentially indicating that adenosine generated from cGAMP rather than ATP is dominant in at least some advanced and metastatic cancers.
ADO signaling and DNA damage responses
DNA damage induced by certain cytotoxic drugs, ultraviolet (UV) light and radiation has been shown to trigger the release of extracellular ATP (4, 5), which can then activate P2X7 receptors and promote inflammation and tumor antigen presentation (4, 16). Damaged mitochondrial DNA can be repaired, but it is most often degraded (127), and autophagy has been shown to restrict the activation of pro-inflammatory pathways by mitochondrial DNA (128). Cytotoxic stress and radiation can also induce the expression of ectonucleotidases capable of converting ATP into immunosuppressive ADO (16, 17, 129).
The upregulation of CD39 and CD73 in response to DNA damage has been linked to the transcriptional activity of cAMP response element-binding protein (CREB) and HIF-1a (130). CREB is phosphorylated by ataxia telangiectasia-mutated (ATM) (131) while HIF-1a activity is induced by DNA-dependent protein kinase (DNA-PK), ATM and ataxia telangiectasia and Rad3-related (ATR) kinase (132–134) (Figure 3).
Figure 3. Crosstalk between DNA damage and adenosine signaling.
DNA damage induces upregulation of CD73 and CD39 ectonucleotidases via CREB and HIF-1. Adenosine produced by CD39 and CD73 signals through A2BR leading to the activation of PKA, PKC and ERK pathways, which have been associated with increased DNA damage responses (DDR). DNA damage also promotes TGF-β signaling, which increases ectonucleotidase expression. EMT enhances DDR and also upregulate CD73, which in turn supports EMT via A2BR signaling.
Hence, DNA damage is often associated with upregulation of ectonucleotidases that favor accumulation of extracellular ADO. In turn, extracellular ADO signaling can prevent DNA damage (135). Activation of p53 can induce A2BR signaling (136). In human and mouse pancreatic tumor cells treated with gemcitabine or radiation, A2BR signaling suppresses accumulation of double strand DNA breaks (137). In human lung cancer and glioblastoma cells, A2BR signaling also improves recovery from radiation-induced DNA damage (138, 139).
Several pathways downstream of A2BR activation are associated with the DDR, which may explain the role of A2BR in regulating DNA damage. For instance, A2BR is the only ADO receptor able to activate both PKA and PKC pathways. On the one hand, PKA cooperates with the DNA damage checkpoint CHK1 to restrain mitotic progression (140). On the other hand, PKC phosphorylates CHK2 and histone H2A after DNA damage and plays a critical role in maintaining DNA integrity (141). PKC signaling is also known to be activated early upon radiation and has been shown to promote radio-resistance (142).
A2BR also cooperates with growth factor signaling pathways (143, 144), which have been implicated in the DDR. Accordingly, DNA damage enhances ligand-induced growth factor receptor signaling (145) and ERK activity upregulates proteins involved in non-homologous end joining (NHEJ) and homologous recombinational repair (HRR) (146) (145). EGFR translocation to the nucleus also occurs following IR and enhances DNA repair through DNA-PKs (147). Recent studies revealed that activation of A2B receptor promotes EGFR translocation to the nucleus in human lung cancer cells treated with radiation and supports DNA repair (139).
Another mechanism that may link ADO signaling to the DRR is epithelial to mesenchymal transition (EMT), a biological program involved in wound healing that contributes to metastasis and radio-resistance (148). Both CD73 and A2BR have been associated with increased EMT (60, 144). In lung cancer cells, A2BR promotes EMT by enhancing ERK signaling (144). Cancer cells that undergo EMT display increase DDR, notably through stabilization of CHK1 by the EMT-inducing transcription factor ZEB1 (149). Ultimately, cancer cells that undergo EMT lose their cell-cell adhesion and acquire migratory and invasive properties, allowing them to enter the bloodstream or lymphatic vessels and metastasize.
Finally, TGF-β is an important paracrine factor that triggers EMT (148, 150) as well as the upregulation of both CD39 and CD73 (151, 152). DNA damage activates TGF-β signaling, which in turn upregulates DDR proteins, including ATM and BRCA1 (153). As such, TGF-β plays a critical role in the repair of double-strand break (154). Whether ADO signaling is involved in this process remains to be investigated.
As indicated above, DNA damage is also associated with activation of the cGAS-STING pathway through the release of cGAMP into the extracellular environment (124, 155). However, tumor cells often overexpress ENPP1, which hydrolases cGAMP and thus inhibits STING signaling (155). ENPP1 hydrolysis of cGAMP, ATP and ADPR also increases extracellular AMP levels, thereby promoting ADO signaling (Figure 4).
Figure 4. Ectonucleotidases inhibit the cGAS-STING pathway.
Upon DNA damage, the cytosolic sensor cGAS produces cyclic GMP-AMP (cGAMP), thereby activating STING and the transcription of type I IFNs and pro-inflammatory cytokines via IRF3 and NF-kB. Cytosolic cGAMP is also released via transporters and gap junctions to activate STING in neighbouring cells. Paracrine activation of STING, notably in antigen presenting cells (APC) and cancer cells, is suppressed by ENPP1 that hydrolyses cGAMP into AMP and GMP. ENPP1 and CD39 further hydrolyses extracellular ATP into AMP. Extracellular AMP is also produced from the conversion of NAD+ to ADPR by CD38, followed by ADPR hydrolysis by ENPP1. Extracellular AMP is hydrolyzed by CD73 into adenosine (ADO), activating cAMP-elevating A2AR and A2BR, which suppresses inflammatory responses, including by blocking NF-kB activity.
In addition to ENPP1, the CD73-ADO axis can also suppress cGAS-STING (137, 156). Our recent study revealed that cancer cell-intrinsic CD73 protected human and mouse pancreatic tumor cells from DNA damage induced by gemcitabine. While A2B receptor signaling partly reversed this phenotype, CD73 may also promote DDR independently of ADO (157). Remarkably, we observed that CD73-deficient tumor cells released significantly greater cGAMP than CD73-expressing tumor cells and upregulated STING target genes (137). Moreover, deletion of cGAS in tumor cells abrogated the in vivo activity of a highly selective CD73 inhibitor (137). Our findings thus highlight a previously unappreciated role for CD73 in regulating the cGAS-STING pathway. Since ENPP1 hydrolyzes both cGAMP and extracellular ATP, co-targeting ENPP1 and CD73 may be synergistic in decreasing ADO signaling and enhancing STING activation. This may be particularly relevant in the context of radiotherapy, known to upregulate both ecto-nucletotidases.
Shifting the balance in favor of immune activation by inhibiting adenosine generation and/or signaling in the TME to improve ICI therapy
Multiple experimental therapeutics targeting CD73, CD39, ENPP1, or the ADO receptors A2A and A2B are in development and some are undergoing clinical testing (158, 159) (Tables 1–3). Strategically, blocking CD73 activity, the final step for ADO generation, should provide the most efficient way to reduce ADO in the TME, whereas targeting CD39 or ENPP1 should enable the accumulation of immune-activating ATP and cGAMP, respectively. For tumors with high levels of adenosine signaling, such as renal cell carcinoma (RCC) and colorectal cancer (160), blocking A2AR may be the most effective intervention. Overall, the choice will have to be guided by the features of the TME and the type of combination therapy that is used. For example, in the context of radiotherapy, blocking CD39 will increase immune activation by ATP, but this effect could be countered by the generation of ADO from NAD+ and cGAMP, which is not dependent on CD39. CD38 plays a role in the alternative pathway of ADO generation, but trials targeting CD38 to intentionally disrupt ADO production have not shown favorable results (108–110), which highlights the need to better understand the complex role of CD38 in immune regulation (118). In addition, given that radiation increases ADO concentration in the TME it may be necessary to block not only A2AR, but also the lower affinity A2B receptor. Below we briefly discuss the results, which have been reported for only a few studies in publications or as meeting abstracts.
Table 1.
Ongoing clinical studies testing ectonucleotidases inhibitors
| Trial | Phase | Title | Disease | Intervention | Responsible Party |
|---|---|---|---|---|---|
| NCT05431270 | Phase 1 | Dose Escalation/Expansion Study of PT 199 (anti- CD73 mAb) Administered Alone and in Combination With a PD-1 Inhibitor | Advanced Solid Tumor | PT199 (Anti-CD73 mAb) | Phanes Therapeutics |
| NCT05270213 | Phase 1 | RBS2418 Evaluation in Subjects With Unresectable or Metastatic Tumors | Advanced Cancer | RBS2418 (ENPP1 antagonist) | Riboscience, LLC. |
| NCT04336098 | Phase 1 | Study of SRF617 in Patients With Advanced Solid Tumors | Advanced Solid Tumor | SRF617 (anti-CD39
mAb) Gemcitabine Nab-Paclitaxel Pembrolizumab |
Surface Oncology |
| NCT04306900 | Phase 1 | TTX-030 in Combination With Immunotherapy and/or Chemotherapy in Subjects With Advanced Cancers | Advanced Solid Tumor | TTX-030 (anti-CD39
mAb) Budigalimab mFOLFOX6, Docetaxel Nab-Paclitaxel Gemcitabine Pembrolizumab |
Trishula Therapeutics, Inc. |
| NCT03884556 | Phase 1 | TTX-030 Single Agent and in Combination With Immunotherapy or Chemotherapy for Patients With Advanced Cancers | Solid Tumor Lymphoma | TTX-030 (anti-CD39
mAb) Pembrolizumab Gemcitabine Nab-Paclitaxel |
Trishula Therapeutics, Inc. |
| NCT03773666 | Phase 1 | A Feasibility Study of Durvalumab +/− Oleclumab as Neoadjuvant Therapy for Muscle-invasive Bladder Cancer (BLASST-2) | Muscle I nvasive Bladder Cancer | Oleclumab (anti-CD73
mAb) Duâlumab |
Dana-Farber Cancer Institute |
| NCT03616886 | Phase 1/2 | Paclitaxel + Carboplatin + Durvalumab With or Without Oleclumabfor Previously Untreated Locally Recurrent Inoperable or Metastatic TNBC (SYNERGY) | Triple Negative Breast Cancer | Oleclumab (anti-CD73
mAb) Paclitaxel Carboplatin Durvalumab |
Jules Bordet Institute |
Table 3.
Ongoing clinical studies testing ectonucleotidases and adenosine receptor inhibitors
| Trial | Phase | Title | Disease | Intervention | Responsible Party |
|---|---|---|---|---|---|
| NCT05177770 | Phase 2 | Study of SRF617 With AB928 (Etrumadenent) and AB122 (Zimberelimab) in Patients With Metastatic Castration Resistant Prostate Cancer | Metastatic castration- resistant prostate cancer | SRF617 (anti-CD39
mAb) AB928 (A2aR/A2bR antagonist) Zimberelimab |
Surface Oncology |
| NCT04381832 | Phase 1/2 | Adenosine Receptor Antagonist Combination Therapy for Metastatic Castrate Resistant Prostate Cancer (ARC-6) | Castration Resistant Prostate Cancer | AB928 (A2aR/A2bR
antagonist) Zimberelimab Quemliclustat (CD73 antagonist) Enzalutamide, Docetaxel Sacituzumab Govitecan |
Arcus Biosciences, Inc. |
| NCT04089553 | Phase 2 | An Open-label, Phase II Study of AZD4635 in Patients With Prostate Cancer | Prostate Cancer | AZD4635 (A2aR
antagonist) Oleclumab (anti-CD73 mAb) Durvalumab |
AstraZeneca |
| NCT04660812 | Phase 1/2 | An Open Label Study Evaluating the Efficacy and Safety of Etrumadenant (AB928) Based Treatment Combinations in Participants With Metastatic Colorectal Cancer (ARC-9) | Metastatic Colorectal Cancer | AB928 (A2aR/A2bR
antagonist) Zimberelimab FOLFOX-6, Bevacizumab Regorafenib Quemliclustat (CD73 antagonist) |
Arcus Biosciences, Inc. |
| NCT03549000 | Phase 1 | A Phase I/Ib Study of NZV930 Alone and in Combination With PDR001 and /or NIR178 in Patients With Advanced Malignancies. | Advanced Solid Tumors | NZV930 (anti-CD73
mAb) PDR001 NIR178/PBF-509 (A2aR antagonist) |
Novartis Pharmaceuticals |
| NCT03454451 | Phase 1 | CPI-006 Alone and in Combination With Ciforadenant and With Pembrolizumab for Patients With Advanced Cancers | Advanced Solid Tumors | CPI-006 (anti-CD73
mAb) Ciforadenant (A2aR antagonist) |
Corvus Pharmaceuticals, Inc. |
Ectonuclotidases inhibitors
Multiple inhibitors of the ectonucleotidase CD73 are undergoing clinical testing, alone or most often in combination with other therapies, including A2A and/or A2B receptor blockade and ICI (Table 1 and 3). Generally, these agents have shown very manageable toxicity but limited activity as monotherapy in early phase studies (161). However, in a randomized study of stage III NSCLC patients treated with chemoradiation, addition of anti-CD73 antibody oleclumab improved overall response rate (ORR) and progression-free survival (PFS) at 12 months over that achieved with anti-PD-L1 alone (162). These results are consistent with preclinical data and suggest that CD73 blockade may be especially effective in the context of DNA damaging therapy and ICI, as discussed above (16, 137). On the other hand, adding oleclumab to durvalumab plus platinum-doublet chemotherapy failed to show clinical benefit in advanced triple-negative breast cancer in a randomized trial, as reported at ESMO 2022 (163). It is still too early to assess the clinical activity of CD39 and ENPP1-targeting agents but results of phase 1 and 1/2 studies are forthcoming (Table 1).
Adenosine signaling inhibitors
Genetic or pharmacologic targeting of A2AR has been shown to improve responses to ICI in several preclinical studies (19, 164, 165). A caveat of A2AR-KO models is that the physiological functions of A2AR during thymic T cell development are also affected (166). In one study, T cell-selective A2AR-KO was shown to compromise the persistence of tumor-specific memory T cells resulting in the inability of mice to control the progression of melanoma and bladder tumors, an effect mediated by reduced IL7 receptor expression on A2AR-KO T cells (167). However, such negative effects have not been observed with the A2AR antagonists that are currently being evaluated in clinical trials, as monotherapy or more often in combination with anti-PD-1/PD-L1 (158, 161). In fact, results of the RCC expansion cohort of NCT02655822 show that A2AR inhibition restored responses to anti-PD-L1 in patients and that durable clinical benefit was associated with increased intratumoral CD8 T cells (168). Inhibition of A2BR was shown to improve the rejection of mouse tumors resistant to A2AR inhibition (169), supporting its therapeutic targeting in the clinic. The simultaneous inhibition of A2A and A2B receptors has the advantage of preventing A2BR-mediated immunosuppression by myeloid cells (169, 170) and disrupting the pro-tumorigenic, tumor-cell intrinsic functions of A2BR discussed above, including the enhancement of radio-resistance. Based on this rationale we have initiated a clinical trial (NCT05024097) testing the dual A2A/A2B receptor antagonist etrumadenant in combination with short course radiotherapy as neoadjuvant treatment in patients with rectal cancer (Table 2). Etrumadenant will be continued during consolidation chemotherapy and anti-PD-1. Other early phase trials are testing adenosine receptor inhibitors with chemoradiation (NCT04892875) in head and neck cancer or in diseases with high adenosine signaling (160).
Table 2.
Ongoing clinical studies testing adenosine receptor inhibitors
| Trial | Phase | Title | Disease | Intervention | Responsible Party |
|---|---|---|---|---|---|
| NCT02403193 | Phase 1 | Trial of PBF-509 and PDR001 in Patients With Advanced Non-small Cell Lung Cancer (NSCLC) (AdenONCO) | NSCLC | PBF-509 (A2aR
antagonist) PDR001 |
Palobiofarma SL |
| NCT05403385 | Phase 2 | Study of Inupadenant (EOS100850) With Chemotherapy as Second Line Treatment for Nonsquamous Non-small Cell Lung Cancer | Metastatic NSCLC Stage III NSCLC | • Inupadenant (A2aR
antagonist) • Placebo • Carboplatin • Pemetrexed |
iTeos Belgium SA |
| NCT03274479 | Phase 1 | PBF-1129 in Patients With NSCLC | Locally Advanced or Metastatic NSCLC | PBF-1129 (A2aR antagonist) | Palobiofarma SL |
| NCT05234307 | Phase 1 | PBF-1129 and Nivolumab for the Treatment of Recurrent or Metastatic Non-Small Cell Lung Cancer | Metastatic NSCLC Recurrent NSCLC | PBF-1129 (A2aR
antagonist) Nivolumab |
The Ohio State University |
| NCT04976660 | Phase 1/2 | TT-4 as a Single Agent in Subjects With Advanced Selected Solid Tumors | Colorectal Cancer Gastric Cancer Hepatocellular Cancer Pancreatic Cancer | TT-4 (A2aR antagonist) | Tarus Therapeutics, Inc. |
| NCT05024097 | Phase 2 | A Phase II Study to Test the Efficacy of AB928 (Dual Adenosine Receptor Antagonist) and AB122 (a PD1 Checkpoint Inhibitor) in Combination With Short Course Radiotherapy and Consolidation Chemotherapy for Rectal Cancer (PANTHER) | Rectal Cancer | AB928 (A2aR/A2bR
antagonist) Radiation Therapy FOLFOX AB122 |
Weill Medical College of Cornell University |
| NCT02655822 | Phase 1/1b | Phase 1/1b Study to Evaluate the Safety and Tolerability of CiforadenantAlone and in Combination With Atezolizumab in Advanced Cancers | Renal Cell Cancer Metastatic Castration Resistant Prostate Cancer | Ciforadenant (A2aR
antagonist) Atezolizumab |
Corvus Pharmaceuticals, Inc. |
| NCT05501054 | Phase 1 b/2 | Phase 1 b/2 Trial of Ipilimumab, Nivolumab, and Ciforadenant (Adenosine A2a Receptor Antagonist) in First- line Advanced Renal Cell Carcinoma. | Renal Cell Carcinoma | • Ipilumumab • Nivolumab • Ciforadenant (A2aR antagonist) |
M D Anderson Cancer Center |
| NCT04969315 | Phase 1/2 | TT-10 as a Single Agent in Subjects With Advanced Selected Solid Tumors | Renal Cell Cancer Castration Resistant Prostate Cancer NSCLC | TT-10 (A2aR antagonist) | Tarus Therapeutics, Inc. |
| NCT04895748 | Phase 1 | DFF332 as a Single Agent and in Combination With Everolimus & Immuno-Oncology Agents in Advanced/Relapsed Renal Cancer & Other Malignancies | Renal Cell Carcinoma | DFF3332, RAD001 PDR001 NIR178/PBF-509 (A2aR antagonist) |
Novartis Pharmaceuticals |
| NCT04892875 | Phase 1 | A Study of Concurrent Chemoradiation in Combination With or Without PD1 Inhibitor AB122 Adenosine 2a Receptor / Adenosine 2b Receptor Inhibitor AB928 Therapies in Locally Advanced Head and Neck Cancers (PANTHEoN) | Head and Neck Cancer | AB122 AB928 (A2aR/A2bR antagonist) Cisplatin, Radiation Therapy |
Vanderbilt-Ingram Cancer Center |
| NCT05198349 | Phase 1 | First in Human Study of M1069 in Advanced Solid Tumors | Metastatic or Locally Advanced Unresectable Solid Tumors | M1069 (A2aR/A2bR antagonist) | EMD Serono |
| NCT04280328 | Phase 1 | Study of Ciforadenant in Combination With Daratumumab in Patients With Relapsed or Refractory Multiple Myeloma | Multiple Myeloma | Ciforadenant (A2aR
antagonist) Daratumumab |
Corvus Pharmaceuticals, Inc. |
| NCT03207867 | Phase 2 | A Phase 2 Study of NIR178 in Combination With PDR001 in Patients With Solid Tumors and Non-Hodgkin Lymphoma | Advanced Solid Tumors NHL | NIR178/PBF-509 (A2aR antagonist), PDR001 | Novartis Pharmaceuticals |
Overall, it is likely that a rationale design of clinical trials based on the evaluation of the tumor TME and careful choice of the combinations tested will identify the clinical settings where inhibition of adenosine generation and/or signaling can provide the most benefits.
Conclusions
DNA damage caused by cytotoxic drugs and radiation can elicit acute inflammation in the tumor microenvironment by at least two processes: i) cell death coupled with release of ATP in the extracellular compartment; ii) the accumulation of DNA in the cytoplasm, leading to activation of cGAS with generation of cGAMP, which can be exported from the cancer cells and activate neighbouring antigen-presenting cells, promoting anti-tumor immunity. However, DNA damaging agents can also induce the expression of ectonucleotidases, such as CD39, CD73 and ENPP1, which convert ATP and cGAMP into ADO. Surface receptors for extracellular ADO are expressed by most immune cells, which explains the broadly immunosuppressive effects of ADO. On the other hand, baseline adenosine signaling is variable in different tumors, which may be a reason for the limited single agent activity of various experimental therapeutics targeting CD73, CD39, ENPP1, and ADO receptors A2A and A2B. In addition, pre-clinical and early clinical data suggest that countering ADO-mediated immunesuppression is often insufficient for tumor rejection without additional stimulation of the immune system. However, this class of immuno-oncology drugs may have a significant impact when used in combination with immune checkpoint inhibitors for the treatment of tumors that are spontaneously enriched in adenosine. We therefore propose that blockade of ADO generation and/or ADO signaling has a key role in improving the anti-tumor immune response when used in combination with DNA damaging therapies, which increase extracellular ADO in the tumor microenvironment.
Recent evidence that ectonucleotidases also regulate the ability of tumor cells to repair DNA further stenghten the rationale for targeting CD73 and possibly other ectonucleotidases in combination with DNA damaging therapies. The choice of targeting strategy must be guided by the unique features of each patient tumor and the specific combination therapies employed.
However, information about which biomarkers are informative is still limited, and more work needs to be done. Rational design of clinical trials, based on the evaluation of the TME and careful selection of combination therapies, will be crucial in identifying the optimal clinical settings for inhibition of adenosine generation and/or signaling to maximize therapeutic benefits.
Acknowledgments
Funding
JS is supported by the Candian Institutes of Health Research and the Fonds de Recherche du Québec - Santé. EG is supported by a Robert A. Winn Diversity in Clinical Trials: Career Development Award from the Bristol Myers Squibb Foundation. SD is supported by NIH/NCI (R01CA201246), the STARR Cancer Consortium (I14-0043), the US Department of Defense Breast Cancer Research Program (#W81XWH1910142), and the Breast Cancer Research Foundation (BCRF-22-053). EW is supported by The Institute of Cancer Research/Royal Marsden (ICR/RM) Cancer Research UK RadNet Centre.
Footnotes
Competing interests
SD has received compensation for consultant/advisory services from Lytix Biopharma, Johnson & Johnson Enterprise Innovation Inc., EMD Serono, Ono Pharmaceutical, and Genentech, and research support from Lytix Biopharma and Boehringer-Ingelheim for unrelated projects.
JS owns stock of Surface Oncology and has received compensation for consultant/advisory services from Surface Oncology and Domain Therapeutics, and research support from Surface Oncology and Domain Therapeutics for unrelated projects. EG receives research support from Arcus Biosciences, Inc. for the PANTHER trial.
The authors declare that they have no competing interests related to this work.
Publisher's Disclaimer: This manuscript has been accepted for publication in Science Immunology. This version has not undergone final editing. Please refer to the complete version of record at www.scienceimmunology.org. The manuscript may not be reproduced or used in any manner that does not fall within the fair use provisions of the Copyright Act without the prior, written permission of AAAS
References
- 1.Sharma P, Allison JP, The future of immune checkpoint therapy. Science 348, 56–61 (2015). [DOI] [PubMed] [Google Scholar]
- 2.Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A, Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 168, 707–723 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Charpentier M, Spada S, Van Nest SJ, Demaria S, Radiation therapy-induced remodeling of the tumor immune microenvironment. Semin Cancer Biol, (2022). [DOI] [PubMed]
- 4.Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, Vermaelen K, Panaretakis T, Mignot G, Ullrich E, Perfettini JL, Schlemmer F, Tasdemir E, Uhl M, Genin P, Civas A, Ryffel B, Kanellopoulos J, Tschopp J, Andre F, Lidereau R, McLaughlin NM, Haynes NM, Smyth MJ, Kroemer G, Zitvogel L, Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med 15, 1170–1178 (2009). [DOI] [PubMed] [Google Scholar]
- 5.Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC, Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 3, e28518 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.McLaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, Harrington KJ, Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer 20, 203–217 (2020). [DOI] [PubMed] [Google Scholar]
- 7.Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, Li XD, Mauceri H, Beckett M, Darga T, Huang X, Gajewski TF, Chen ZJ, Fu YX, Weichselbaum RR, STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 41, 843–852 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Carozza JA, Böhnert V, Nguyen KC, Skariah G, Shaw KE, Brown JA, Rafat M, von Eyben R, Graves EE, Glenn JS, Smith M, Li L, Extracellular cGAMP is a cancer-cell-produced immunotransmitter involved in radiation-induced anticancer immunity. Nature Cancer 1, 184–196 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Demaria S, Formenti SC, The abscopal effect 67 years later: from a side story to center stage. Br J Radiol 93, 20200042 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Formenti SC, Rudqvist NP, Golden E, Cooper B, Wennerberg E, Lhuillier C, Vanpouille-Box C, Friedman K, Ferrari de Andrade L, Wucherpfennig KW, Heguy A, Imai N, Gnjatic S, Emerson RO, Zhou XK, Zhang T, Chachoua A, Demaria S, Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat Med 24, 1845–1851 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Parikh AR, Szabolcs A, Allen JN, Clark JW, Wo JY, Raabe M, Thel H, Hoyos D, Mehta A, Arshad S, Lieb DJ, Drapek LC, Blaszkowsky LS, Giantonio BJ, Weekes CD, Zhu AX, Goyal L, Nipp RD, Dubois JS, Van Seventer EE, Foreman BE, Matlack LE, Ly L, Meurer JA, Hacohen N, Ryan DP, Yeap BY, Corcoran RB, Greenbaum BD, Ting DT, Hong TS, Radiation therapy enhances immunotherapy response in microsatellite stable colorectal and pancreatic adenocarcinoma in a phase II trial. Nat Cancer 2, 1124–1135 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Altorki NK, McGraw TE, Borczuk AC, Saxena A, Port JL, Stiles BM, Lee BE, Sanfilippo NJ, Scheff RJ, Pua BB, Gruden JF, Christos PJ, Spinelli C, Gakuria J, Uppal M, Binder B, Elemento O, Ballman KV, Formenti SC, Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial. Lancet Oncol 22, 824–835 (2021). [DOI] [PubMed] [Google Scholar]
- 13.Leidner R, Crittenden M, Young K, Xiao H, Wu Y, Couey MA, Patel AA, Cheng AC, Watters AL, Bifulco C, Morris G, Rushforth L, Nemeth S, Urba WJ, Gough M, Bell RB, Neoadjuvant immunoradiotherapy results in high rate of complete pathological response and clinical to pathological downstaging in locally advanced head and neck squamous cell carcinoma. J Immunother Cancer 9, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Pointer KB, Pitroda SP, Weichselbaum RR, Radiotherapy and immunotherapy: open questions and future strategies. Trends Cancer 8, 9–20 (2022). [DOI] [PubMed] [Google Scholar]
- 15.Demaria S, Guha C, Schoenfeld J, Morris Z, Monjazeb A, Sikora A, Crittenden M, Shiao S, Khleif S, Gupta S, Formenti SC, Vikram B, Coleman CN, Ahmed MM, Radiation dose and fraction in immunotherapy: one-size regimen does not fit all settings, so how does one choose? J Immunother Cancer 9, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wennerberg E, Spada S, Rudqvist NP, Lhuillier C, Gruber S, Chen Q, Zhang F, Zhou XK, Gross SS, Formenti SC, Demaria S, CD73 Blockade Promotes Dendritic Cell Infiltration of Irradiated Tumors and Tumor Rejection. Cancer Immunol Res 8, 465–478 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Tsukui H, Horie H, Koinuma K, Ohzawa H, Sakuma Y, Hosoya Y, Yamaguchi H, Yoshimura K, Lefor AK, Sata N, Kitayama J, CD73 blockade enhances the local and abscopal effects of radiotherapy in a murine rectal cancer model. BMC Cancer 20, 411 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Puchalowicz K, Tarnowski M, Baranowska-Bosiacka I, Chlubek D, Dziedziejko V, P2X and P2Y receptors-role in the pathophysiology of the nervous system. Int J Mol Sci 15, 23672–23704 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ohta A, Gorelik E, Prasad SJ, Ronchese F, Lukashev D, Wong MK, Huang X, Caldwell S, Liu K, Smith P, Chen JF, Jackson EK, Apasov S, Abrams S, Sitkovsky M, A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci U S A 103, 13132–13137 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Vijayan D, Young A, Teng MWL, Smyth MJ, Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer 17, 709–724 (2017). [DOI] [PubMed] [Google Scholar]
- 21.Pellegatti P, Raffaghello L, Bianchi G, Piccardi F, Pistoia V, Di Virgilio F, Increased level of extracellular ATP at tumor sites: in vivo imaging with plasma membrane luciferase. PLoS One 3, e2599 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Di Virgilio F, Dal Ben D, Sarti AC, Giuliani AL, Falzoni S, The P2X7 Receptor in Infection and Inflammation. Immunity 47, 15–31 (2017). [DOI] [PubMed] [Google Scholar]
- 23.Silverman WR, de Rivero Vaccari JP, Locovei S, Qiu F, Carlsson SK, Scemes E, Keane RW, Dahl G, The pannexin 1 channel activates the inflammasome in neurons and astrocytes. J Biol Chem 284, 18143–18151 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Eltzschig HK, Eckle T, Mager A, Kuper N, Karcher C, Weissmuller T, Boengler K, Schulz R, Robson SC, Colgan SP, ATP release from activated neutrophils occurs via connexin 43 and modulates adenosine-dependent endothelial cell function. Circ Res 99, 1100–1108 (2006). [DOI] [PubMed] [Google Scholar]
- 25.Vultaggio-Poma V, Falzoni S, Chiozzi P, Sarti AC, Adinolfi E, Giuliani AL, Sanchez-Melgar A, Boldrini P, Zanoni M, Tesei A, Pinton P, Di Virgilio F, Extracellular ATP is increased by release of ATP-loaded microparticles triggered by nutrient deprivation. Theranostics 12, 859–874 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Feng M, Elaib Z, Borgel D, Denis CV, Adam F, Bryckaert M, Rosa JP, Bobe R, NAADP/SERCA3-Dependent Ca(2+) Stores Pathway Specifically Controls Early Autocrine ADP Secretion Potentiating Platelet Activation. Circ Res 127, e166–e183 (2020). [DOI] [PubMed] [Google Scholar]
- 27.Fredholm BB, AP IJ, Jacobson KA, Linden J, Muller CE, International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine receptors--an update. Pharmacol Rev 63, 1–34 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kaczmarek E, Koziak K, Sevigny J, Siegel JB, Anrather J, Beaudoin AR, Bach FH, Robson SC, Identification and characterization of CD39/vascular ATP diphosphohydrolase. J Biol Chem 271, 33116–33122 (1996). [DOI] [PubMed] [Google Scholar]
- 29.Knapp K, Zebisch M, Pippel J, El-Tayeb A, Muller CE, Strater N, Crystal structure of the human ecto-5’-nucleotidase (CD73): insights into the regulation of purinergic signaling. Structure 20, 2161–2173 (2012). [DOI] [PubMed] [Google Scholar]
- 30.Kato K, Nishimasu H, Oikawa D, Hirano S, Hirano H, Kasuya G, Ishitani R, Tokunaga F, Nureki O, Structural insights into cGAMP degradation by Ecto-nucleotide pyrophosphatase phosphodiesterase 1. Nat Commun 9, 4424 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Martins I, Wang Y, Michaud M, Ma Y, Sukkurwala AQ, Shen S, Kepp O, Metivier D, Galluzzi L, Perfettini JL, Zitvogel L, Kroemer G, Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ 21, 79–91 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, Chan TA, Coukos G, Demaria S, Deutsch E, Draganov D, Edelson RL, Formenti SC, Fucikova J, Gabriele L, Gaipl US, Gameiro SR, Garg AD, Golden E, Han J, Harrington KJ, Hemminki A, Hodge JW, Hossain DMS, Illidge T, Karin M, Kaufman HL, Kepp O, Kroemer G, Lasarte JJ, Loi S, Lotze MT, Manic G, Merghoub T, Melcher AA, Mossman KL, Prosper F, Rekdal O, Rescigno M, Riganti C, Sistigu A, Smyth MJ, Spisek R, Stagg J, Strauss BE, Tang D, Tatsuno K, van Gool SW, Vandenabeele P, Yamazaki T, Zamarin D, Zitvogel L, Cesano A, Marincola FM, Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer 8, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kroemer G, Galassi C, Zitvogel L, Galluzzi L, Immunogenic cell stress and death. Nat Immunol 23, 487–500 (2022). [DOI] [PubMed] [Google Scholar]
- 34.Ma Z, Li W, Yoshiya S, Xu Y, Hata M, El-Darawish Y, Markova T, Yamanishi K, Yamanishi H, Tahara H, Tanaka Y, Okamura H, Augmentation of Immune Checkpoint Cancer Immunotherapy with IL18. Clin Cancer Res 22, 2969–2980 (2016). [DOI] [PubMed] [Google Scholar]
- 35.Li XY, Moesta AK, Xiao C, Nakamura K, Casey M, Zhang H, Madore J, Lepletier A, Aguilera AR, Sundarrajan A, Jacoberger-Foissac C, Wong C, Dela Cruz T, Welch M, Lerner AG, Spatola BN, Soros VB, Corbin J, Anderson AC, Effern M, Holzel M, Robson SC, Johnston RL, Waddell N, Smith C, Bald T, Geetha N, Beers C, Teng MWL, Smyth MJ, Targeting CD39 in Cancer Reveals an Extracellular ATP- and Inflammasome-Driven Tumor Immunity. Cancer Discov 9, 1754–1773 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, Park D, Woodson RI, Ostankovich M, Sharma P, Lysiak JJ, Harden TK, Leitinger N, Ravichandran KS, Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 461, 282–286 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hanley PJ, Musset B, Renigunta V, Limberg SH, Dalpke AH, Sus R, Heeg KM, Preisig-Muller R, Daut J, Extracellular ATP induces oscillations of intracellular Ca2+ and membrane potential and promotes transcription of IL-6 in macrophages. Proc Natl Acad Sci U S A 101, 9479–9484 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cohen HB, Briggs KT, Marino JP, Ravid K, Robson SC, Mosser DM, TLR stimulation initiates a CD39-based autoregulatory mechanism that limits macrophage inflammatory responses. Blood 122, 1935–1945 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Layhadi JA, Turner J, Crossman D, Fountain SJ, ATP Evokes Ca(2+) Responses and CXCL5 Secretion via P2X(4) Receptor Activation in Human Monocyte-Derived Macrophages. J Immunol 200, 1159–1168 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Savio LEB, de Andrade Mello P, Figliuolo VR, de Avelar Almeida TF, Santana PT, Oliveira SDS, Silva CLM, Feldbrugge L, Csizmadia E, Minshall RD, Longhi MS, Wu Y, Robson SC, Coutinho-Silva R, CD39 limits P2X7 receptor inflammatory signaling and attenuates sepsis-induced liver injury. J Hepatol 67, 716–726 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kayagaki N, Stowe IB, Lee BL, O’Rourke K, Anderson K, Warming S, Cuellar T, Haley B, Roose-Girma M, Phung QT, Liu PS, Lill JR, Li H, Wu J, Kummerfeld S, Zhang J, Lee WP, Snipas SJ, Salvesen GS, Morris LX, Fitzgerald L, Zhang Y, Bertram EM, Goodnow CC, Dixit VM, Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671 (2015). [DOI] [PubMed] [Google Scholar]
- 42.Wang Q, Wang Y, Ding J, Wang C, Zhou X, Gao W, Huang H, Shao F, Liu Z, A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature 579, 421–426 (2020). [DOI] [PubMed] [Google Scholar]
- 43.Ledderose C, Liu K, Kondo Y, Slubowski CJ, Dertnig T, Denicolo S, Arbab M, Hubner J, Konrad K, Fakhari M, Lederer JA, Robson SC, Visner GA, Junger WG, Purinergic P2X4 receptors and mitochondrial ATP production regulate T cell migration. J Clin Invest 128, 3583–3594 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Brock VJ, Wolf IMA, Er-Lukowiak M, Lory N, Stahler T, Woelk LM, Mittrucker HW, Muller CE, Koch-Nolte F, Rissiek B, Werner R, Guse AH, Diercks BP, P2X4 and P2X7 are essential players in basal T cell activity and Ca(2+) signaling milliseconds after T cell activation. Sci Adv 8, eabl9770 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Mutini C, Falzoni S, Ferrari D, Chiozzi P, Morelli A, Baricordi OR, Collo G, Ricciardi-Castagnoli P, Di Virgilio F, Mouse dendritic cells express the P2X7 purinergic receptor: characterization and possible participation in antigen presentation. J Immunol 163, 1958–1965 (1999). [PubMed] [Google Scholar]
- 46.Borges da Silva H, Beura LK, Wang H, Hanse EA, Gore R, Scott MC, Walsh DA, Block KE, Fonseca R, Yan Y, Hippen KL, Blazar BR, Masopust D, Kelekar A, Vulchanova L, Hogquist KA, Jameson SC, The purinergic receptor P2RX7 directs metabolic fitness of long-lived memory CD8(+) T cells. Nature 559, 264–268 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Borges da Silva H, Peng C, Wang H, Wanhainen KM, Ma C, Lopez S, Khoruts A, Zhang N, Jameson SC, Sensing of ATP via the Purinergic Receptor P2RX7 Promotes CD8(+) Trm Cell Generation by Enhancing Their Sensitivity to the Cytokine TGF-beta. Immunity 53, 158–171 e156 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Romagnani A, Rottoli E, Mazza EMC, Rezzonico-Jost T, De Ponte Conti B, Proietti M, Perotti M, Civanelli E, Perruzza L, Catapano AL, Baragetti A, Tenedini E, Tagliafico E, Falzoni S, Di Virgilio F, Norata GD, Bicciato S, Grassi F, P2X7 Receptor Activity Limits Accumulation of T Cells within Tumors. Cancer Res 80, 3906–3919 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Schenk U, Frascoli M, Proietti M, Geffers R, Traggiai E, Buer J, Ricordi C, Westendorf AM, Grassi F, ATP inhibits the generation and function of regulatory T cells through the activation of purinergic P2X receptors. Sci Signal 4, ra12 (2011). [DOI] [PubMed] [Google Scholar]
- 50.Faliti CE, Gualtierotti R, Rottoli E, Gerosa M, Perruzza L, Romagnani A, Pellegrini G, De Ponte Conti B, Rossi RL, Idzko M, Mazza EMC, Bicciato S, Traggiai E, Meroni PL, Grassi F, P2X7 receptor restrains pathogenic Tfh cell generation in systemic lupus erythematosus. J Exp Med 216, 317–336 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cao W, Fang F, Gould T, Li X, Kim C, Gustafson C, Lambert S, Weyand CM, Goronzy JJ, Ecto-NTPDase CD39 is a negative checkpoint that inhibits follicular helper cell generation. J Clin Invest 130, 3422–3436 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Baroja-Mazo A, Penin-Franch A, Lucas-Ruiz F, de Torre-Minguela C, Alarcon-Vila C, Hernandez-Caselles T, Pelegrin P, P2X7 receptor activation impairs antitumour activity of natural killer cells. Br J Pharmacol 180, 111–128 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Adinolfi E, Capece M, Franceschini A, Falzoni S, Giuliani AL, Rotondo A, Sarti AC, Bonora M, Syberg S, Corigliano D, Pinton P, Jorgensen NR, Abelli L, Emionite L, Raffaghello L, Pistoia V, Di Virgilio F, Accelerated tumor progression in mice lacking the ATP receptor P2X7. Cancer Res 75, 635–644 (2015). [DOI] [PubMed] [Google Scholar]
- 54.Pippel A, Bessler B, Klapperstuck M, Markwardt F, Inhibition of antigen receptor-dependent Ca(2+) signals and NF-AT activation by P2X7 receptors in human B lymphocytes. Cell Calcium 57, 275–289 (2015). [DOI] [PubMed] [Google Scholar]
- 55.Figueiro F, Muller L, Funk S, Jackson EK, Battastini AM, Whiteside TL , Phenotypic and functional characteristics of CD39(high) human regulatory B cells (Breg). Oncoimmunology 5, e1082703 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Jeske SS, Brand M, Ziebart A, Laban S, Doescher J, Greve J, Jackson EK, Hoffmann TK, Brunner C, Schuler PJ, Adenosine-producing regulatory B cells in head and neck cancer. Cancer Immunol Immunother 69, 1205–1216 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nascimento DC, Viacava PR, Ferreira RG, Damaceno MA, Pineros AR, Melo PH, Donate PB, Toller-Kawahisa JE, Zoppi D, Veras FP, Peres RS, Menezes-Silva L, Caetite D, Oliveira AER, Castro IMS, Kauffenstein G, Nakaya HI, Borges MC, Zamboni DS, Fonseca DM, Paschoal JAR, Cunha TM, Quesniaux V, Linden J, Cunha FQ, Ryffel B, Alves-Filho JC, Sepsis expands a CD39(+) plasmablast population that promotes immunosuppression via adenosine-mediated inhibition of macrophage antimicrobial activity. Immunity 54, 2024–2041 e2028 (2021). [DOI] [PubMed] [Google Scholar]
- 58.Bullen JW, Tchernyshyov I, Holewinski RJ, DeVine L, Wu F, Venkatraman V, Kass DL, Cole RN, Van Eyk J, Semenza GL, Protein kinase A-dependent phosphorylation stimulates the transcriptional activity of hypoxia-inducible factor 1. Sci Signal 9, ra56 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Reinhardt J, Landsberg J, Schmid-Burgk JL, Ramis BB, Bald T, Glodde N, Lopez-Ramos D, Young A, Ngiow SF, Nettersheim D, Schorle H, Quast T, Kolanus W, Schadendorf D, Long GV, Madore J, Scolyer RA, Ribas A, Smyth MJ, Tumeh PC, Tuting T, Holzel M, MAPK Signaling and Inflammation Link Melanoma Phenotype Switching to Induction of CD73 during Immunotherapy. Cancer Res 77, 4697–4709 (2017). [DOI] [PubMed] [Google Scholar]
- 60.Lupia M, Angiolini F, Bertalot G, Freddi S, Sachsenmeier KF, Chisci E, Kutryb-Zajac B, Confalonieri S, Smolenski RT, Giovannoni R, Colombo N, Bianchi F, Cavallaro U, CD73 Regulates Stemness and Epithelial-Mesenchymal Transition in Ovarian Cancer-Initiating Cells. Stem Cell Reports 10, 1412–1425 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Han Y, Lee T, He Y, Raman R, Irizarry A, Martin ML, Giaccone G, The regulation of CD73 in non-small cell lung cancer. Eur J Cancer 170, 91–102 (2022). [DOI] [PubMed] [Google Scholar]
- 62.Allard B, Longhi MS, Robson SC, Stagg J, The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev 276, 121–144 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ohta A, Sitkovsky M, Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 414, 916–920 (2001). [DOI] [PubMed] [Google Scholar]
- 64.Welihinda AA, Kaur M, Greene K, Zhai Y, Amento EP, The adenosine metabolite inosine is a functional agonist of the adenosine A2A receptor with a unique signaling bias. Cell Signal 28, 552–560 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Mager LF, Burkhard R, Pett N, Cooke NCA, Brown K, Ramay H, Paik S, Stagg J, Groves RA, Gallo M, Lewis IA, Geuking MB, McCoy KD, Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 369, 1481–1489 (2020). [DOI] [PubMed] [Google Scholar]
- 66.Chen S, Akdemir I, Fan J, Linden J, Zhang B, Cekic C, The Expression of Adenosine A2B Receptor on Antigen-Presenting Cells Suppresses CD8(+) T-cell Responses and Promotes Tumor Growth. Cancer Immunol Res 8, 1064–1074 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Borodovsky A, Barbon CM, Wang Y, Ye M, Prickett L, Chandra D, Shaw J, Deng N, Sachsenmeier K, Clarke JD, Linghu B, Brown GA, Brown J, Congreve M, Cheng RK, Dore AS, Hurrell E, Shao W, Woessner R, Reimer C, Drew L, Fawell S, Schuller AG, Mele DA, Small molecule AZD4635 inhibitor of A(2A)R signaling rescues immune cell function including CD103(+) dendritic cells enhancing anti-tumor immunity. J Immunother Cancer 8, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wilson JM, Ross WG, Agbai ON, Frazier R, Figler RA, Rieger J, Linden J, Ernst PB, The A2B adenosine receptor impairs the maturation and immunogenicity of dendritic cells. J Immunol 182, 4616–4623 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Giuffrida L, Sek K, Henderson MA, Lai J, Chen AXY, Meyran D, Todd KL, Petley EV, Mardiana S, Molck C, Stewart GD, Solomon BJ, Parish IA, Neeson PJ, Harrison SJ, Kats LM, House IG, Darcy PK, Beavis PA, CRISPR/Cas9 mediated deletion of the adenosine A2A receptor enhances CAR T cell efficacy. Nat Commun 12, 3236 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Allard B, Pommey S, Smyth MJ, Stagg J, Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin Cancer Res 19, 5626–5635 (2013). [DOI] [PubMed] [Google Scholar]
- 71.Schneider E, Winzer R, Rissiek A, Ricklefs I, Meyer-Schwesinger C, Ricklefs FL, Bauche A, Behrends J, Reimer R, Brenna S, Wasielewski H, Lauten M, Rissiek B, Puig B, Cortesi F, Magnus T, Fliegert R, Muller CE, Gagliani N, Tolosa E, CD73-mediated adenosine production by CD8 T cell-derived extracellular vesicles constitutes an intrinsic mechanism of immune suppression. Nat Commun 12, 5911 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Fang F, Cao W, Zhu W, Lam N, Li L, Gaddam S, Wang Y, Kim C, Lambert S, Zhang H, Hu B, Farber DL, Weyand CM, Goronzy JJ, The cell-surface 5’-nucleotidase CD73 defines a functional T memory cell subset that declines with age. Cell Rep 37, 109981 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Fang F, Cao W, Mu Y, Okuyama H, Li L, Qiu J, Weyand CM, Goronzy JJ, IL-4 prevents adenosine-mediated immunoregulation by inhibiting CD39 expression. JCI Insight 7, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Simoni Y, Becht E, Fehlings M, Loh CY, Koo SL, Teng KWW, Yeong JPS, Nahar R, Zhang T, Kared H, Duan K, Ang N, Poidinger M, Lee YY, Larbi A, Khng AJ, Tan E, Fu C, Mathew R, Teo M, Lim WT, Toh CK, Ong BH, Koh T, Hillmer AM, Takano A, Lim TKH, Tan EH, Zhai W, Tan DSW, Tan IB, Newell EW, Bystander CD8(+) T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 557, 575–579 (2018). [DOI] [PubMed] [Google Scholar]
- 75.Gupta PK, Godec J, Wolski D, Adland E, Yates K, Pauken KE, Cosgrove C, Ledderose C, Junger WG, Robson SC, Wherry EJ, Alter G, Goulder PJ, Klenerman P, Sharpe AH, Lauer GM, Haining WN, CD39 Expression Identifies Terminally Exhausted CD8+ T Cells. PLoS Pathog 11, e1005177 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Schuler PJ, Saze Z, Hong CS, Muller L, Gillespie DG, Cheng D, Harasymczuk M, Mandapathil M, Lang S, Jackson EK, Whiteside TL, Human CD4+ CD39+ regulatory T cells produce adenosine upon co-expression of surface CD73 or contact with CD73+ exosomes or CD73+ cells. Clin Exp Immunol 177, 531–543 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Grignano MA, Bruno S, Viglio S, Avanzini MA, Tapparo M, Ramus M, Croce S, Valsecchi C, Pattonieri EF, Ceccarelli G, Manzoni F, Asti A, Libetta C, Sepe V, Iadarola P, Gregorini M, Rampino T, CD73-Adenosinergic Axis Mediates the Protective Effect of Extracellular Vesicles Derived from Mesenchymal Stromal Cells on Ischemic Renal Damage in a Rat Model of Donation after Circulatory Death. Int J Mol Sci 23, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ohta A, Kini R, Ohta A, Subramanian M, Madasu M, Sitkovsky M, The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front Immunol 3, 190 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Muhammad F, Wang D, Montieth A, Lee S, Preble J, Foster CS, Larson TA, Ding K, Dvorak JD, Lee DJ, PD-1(+) melanocortin receptor dependent-Treg cells prevent autoimmune disease. Sci Rep 9, 16941 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kinsey GR, Huang L, Jaworska K, Khutsishvili K, Becker DA, Ye H, Lobo PI, Okusa MD, Autocrine adenosine signaling promotes regulatory T cell-mediated renal protection. J Am Soc Nephrol 23, 1528–1537 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Gerner MC, Ziegler LS, Schmidt RLJ, Krenn M, Zimprich F, Uyanik-Unal K, Konstantopoulou V, Derdak S, Del Favero G, Schwarzinger I, Boztug K, Schmetterer KG, The TGF-b/SOX4 axis and ROS-driven autophagy co-mediate CD39 expression in regulatory T-cells. FASEB J 34, 8367–8384 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Jarvis LB, Rainbow DB, Coppard V, Howlett SK, Georgieva Z, Davies JL, Mullay HK, Hester J, Ashmore T, Van Den Bosch A, Grist JT, Coles AJ, Mousa HS, Pluchino S, Mahbubani KT, Griffin JL, Saeb-Parsy K, Issa F, Peruzzotti-Jametti L, Wicker LS, Jones JL, Therapeutically expanded human regulatory T-cells are super-suppressive due to HIF1A induced expression of CD73. Commun Biol 4, 1186 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sim GC, Martin-Orozco N, Jin L, Yang Y, Wu S, Washington E, Sanders D, Lacey C, Wang Y, Vence L, Hwu P, Radvanyi L, IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J Clin Invest 124, 99–110 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Young A, Ngiow SF, Gao Y, Patch AM, Barkauskas DS, Messaoudene M, Lin G, Coudert JD, Stannard KA, Zitvogel L, Degli-Esposti MA, Vivier E, Waddell N, Linden J, Huntington ND, Souza-Fonseca-Guimaraes F, Smyth MJ, A2AR Adenosine Signaling Suppresses Natural Killer Cell Maturation in the Tumor Microenvironment. Cancer Res 78, 1003–1016 (2018). [DOI] [PubMed] [Google Scholar]
- 85.Terme M, Ullrich E, Aymeric L, Meinhardt K, Coudert JD, Desbois M, Ghiringhelli F, Viaud S, Ryffel B, Yagita H, Chen L, Mecheri S, Kaplanski G, Prevost-Blondel A, Kato M, Schultze JL, Tartour E, Kroemer G, Degli-Esposti M, Chaput N, Zitvogel L, Cancer-induced immunosuppression: IL-18-elicited immunoablative NK cells. Cancer Res 72, 2757–2767 (2012). [DOI] [PubMed] [Google Scholar]
- 86.Neo SY, Yang Y, Record J, Ma R, Chen X, Chen Z, Tobin NP, Blake E, Seitz C, Thomas R, Wagner AK, Andersson J, de Boniface J, Bergh J, Murray S, Alici E, Childs R, Johansson M, Westerberg LS, Haglund F, Hartman J, Lundqvist A, CD73 immune checkpoint defines regulatory NK cells within the tumor microenvironment. J Clin Invest 130, 1185–1198 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Saze Z, Schuler PJ, Hong CS, Cheng D, Jackson EK, Whiteside TL, Adenosine production by human B cells and B cell-mediated suppression of activated T cells. Blood 122, 9–18 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Willingham SB, Criner G, Hill C, Hu S, Rudnick JA, Daine-Matsuoka B, Hsieh J, Mashhedi H, Hotson AN, Brody J, Marron T, Piccione E, Buggy JJ, Mahabhashyam S, Jones WB, Mobasher M, Miller RA, Characterization and Phase 1 Trial of a B Cell Activating Anti-CD73 Antibody for the Immunotherapy of COVID-19. medRxiv, (2020).
- 89.Resta R, Yamashita Y, Thompson LF, Ecto-enzyme and signaling functions of lymphocyte CD73. Immunol Rev 161, 95–109 (1998). [DOI] [PubMed] [Google Scholar]
- 90.Vences-Catalan F, Santos-Argumedo L, CD38 through the life of a murine B lymphocyte. IUBMB Life 63, 840–846 (2011). [DOI] [PubMed] [Google Scholar]
- 91.Reinherz EL, Kung PC, Goldstein G, Levey RH, Schlossman SF, Discrete stages of human intrathymic differentiation: analysis of normal thymocytes and leukemic lymphoblasts of T-cell lineage. Proc Natl Acad Sci U S A 77, 1588–1592 (1980). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Aksoy P, White TA, Thompson M, Chini EN, Regulation of intracellular levels of NAD: a novel role for CD38. Biochem Biophys Res Commun 345, 1386–1392 (2006). [DOI] [PubMed] [Google Scholar]
- 93.Corda D, Di Girolamo M, Functional aspects of protein mono-ADP-ribosylation. EMBO J 22, 1953–1958 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Hassa PO, Hottiger MO, The diverse biological roles of mammalian PARPS, a small but powerful family of poly-ADP-ribose polymerases. Front Biosci 13, 3046–3082 (2008). [DOI] [PubMed] [Google Scholar]
- 95.Covarrubias AJ, Perrone R, Grozio A, Verdin E, NAD(+) metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cell Biol 22, 119–141 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Morandi F, Morandi B, Horenstein AL, Chillemi A, Quarona V, Zaccarello G, Carrega P, Ferlazzo G, Mingari MC, Moretta L, Pistoia V, Malavasi F, A non-canonical adenosinergic pathway led by CD38 in human melanoma cells induces suppression of T cell proliferation. Oncotarget 6, 25602–25618 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Horenstein AL, Chillemi A, Zaccarello G, Bruzzone S, Quarona V, Zito A, Serra S, Malavasi F, A CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology 2, e26246 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.van de Donk N, Richardson PG, Malavasi F, CD38 antibodies in multiple myeloma: back to the future. Blood 131, 13–29 (2018). [DOI] [PubMed] [Google Scholar]
- 99.Horenstein AL, Chillemi A, Quarona V, Zito A, Roato I, Morandi F, Marimpietri D, Bolzoni M, Toscani D, Oldham RJ, Cuccioloni M, Sasser AK, Pistoia V, Giuliani N, Malavasi F, NAD(+)-Metabolizing Ectoenzymes in Remodeling Tumor-Host Interactions: The Human Myeloma Model. Cells 4, 520–537 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Chen L, Diao L, Yang Y, Yi X, Rodriguez BL, Li Y, Villalobos PA, Cascone T, Liu X, Tan L, Lorenzi PL, Huang A, Zhao Q, Peng D, Fradette JJ, Peng DH, Ungewiss C, Roybal J, Tong P, Oba J, Skoulidis F, Peng W, Carter BW, Gay CM, Fan Y, Class CA, Zhu J, Rodriguez-Canales J, Kawakami M, Byers LA, Woodman SE, Papadimitrakopoulou VA, Dmitrovsky E, Wang J, Ullrich SE, Wistuba II, Heymach JV, Qin FX, Gibbons DL, CD38-Mediated Immunosuppression as a Mechanism of Tumor Cell Escape from PD-1/PD-L1 Blockade. Cancer Discov 8, 1156–1175 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Guo C, Crespo M, Gurel B, Dolling D, Rekowski J, Sharp A, Petremolo A, Sumanasuriya S, Rodrigues DN, Ferreira A, Pereira R, Figueiredo I, Mehra N, Lambros MBK, Neeb A, Gil V, Seed G, Terstappen L, Alimonti A, Drake CG, Yuan W, de Bono JS, International SUCPCFPCDT, CD38 in Advanced Prostate Cancers. Eur Urol 79, 736–746 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Chmielewski JP, Bowlby SC, Wheeler FB, Shi L, Sui G, Davis AL, Howard TD, D’Agostino RB Jr., Miller LD, Sirintrapun SJ, Cramer SD, Kridel SJ, CD38 Inhibits Prostate Cancer Metabolism and Proliferation by Reducing Cellular NAD(+) Pools. Mol Cancer Res 16, 1687–1700 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Karakasheva TA, Waldron TJ, Eruslanov E, Kim SB, Lee JS, O’Brien S, Hicks PD, Basu D, Singhal S, Malavasi F, Rustgi AK, CD38-Expressing Myeloid-Derived Suppressor Cells Promote Tumor Growth in a Murine Model of Esophageal Cancer. Cancer Res 75, 4074–4085 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lam JH, Ng HHM, Lim CJ, Sim XN, Malavasi F, Li H, Loh JJH, Sabai K, Kim JK, Ong CCH, Loh T, Leow WQ, Choo SP, Toh HC, Lee SY, Chan CY, Chew V, Lim TS, Yeong J, Lim TKH, Expression of CD38 on Macrophages Predicts Improved Prognosis in Hepatocellular Carcinoma. Front Immunol 10, 2093 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Ng HHM, Lee RY, Goh S, Tay ISY, Lim X, Lee B, Chew V, Li H, Tan B, Lim S, Lim JCT, Au B, Loh JJH, Saraf S, Connolly JE, Loh T, Leow WQ, Lee JJX, Toh HC, Malavasi F, Lee SY, Chow P, Newell EW, Choo SP, Tai D, Yeong J, Lim TKH, Immunohistochemical scoring of CD38 in the tumor microenvironment predicts responsiveness to anti-PD-1/PD-L1 immunotherapy in hepatocellular carcinoma. J Immunother Cancer 8, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Drach J, McQueen T, Engel H, Andreeff M, Robertson KA, Collins SJ, Malavasi F, Mehta K, Retinoic acid-induced expression of CD38 antigen in myeloid cells is mediated through retinoic acid receptor-alpha. Cancer Res 54, 1746–1752 (1994). [PubMed] [Google Scholar]
- 107.Karakasheva TA, Dominguez GA, Hashimoto A, Lin EW, Chiu C, Sasser K, Lee JW, Beatty GL, Gabrilovich DI, Rustgi AK, CD38+ M-MDSC expansion characterizes a subset of advanced colorectal cancer patients. JCI Insight 3, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Simonelli M, Garralda E, Eskens F, Gil-Martin M, Yen CJ, Obermannova R, Chao Y, Lonardi S, Melichar B, Moreno V, Yu ML, Bongiovanni A, Calvo E, Rottey S, Machiels JP, Gonzalez-Martin A, Paz-Ares L, Chang CL, Mason W, Lin CC, Reardon DA, Vieito M, Santoro A, Meng R, Abbadessa G, Menas F, Lee H, Liu Q, Combeau C, Ternes N, Ziti-Ljajic S, Massard C, Isatuximab plus atezolizumab in patients with advanced solid tumors: results from a phase I/II, open-label, multicenter study. ESMO Open 7, 100562 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Zucali PA, Lin CC, Carthon BC, Bauer TM, Tucci M, Italiano A, Iacovelli R, Su WC, Massard C, Saleh M, Daniele G, Greystoke A, Gutierrez M, Pant S, Shen YC, Perrino M, Meng R, Abbadessa G, Lee H, Dong Y, Chiron M, Wang R, Loumagne L, Lepine L, de Bono J, Targeting CD38 and PD-1 with isatuximab plus cemiplimab in patients with advanced solid malignancies: results from a phase I/II open-label, multicenter study. J Immunother Cancer 10, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Pillai RN, Ramalingam SS, Thayu M, Lorenzini P, Alvarez Arias DA, Moy C, Hutnick N, Knoblauch R, Feng H, Kane C, Horn L, Reck M, Ponce S, Investigators LUC, Daratumumab Plus Atezolizumab in Previously Treated Advanced or Metastatic NSCLC: Brief Report on a Randomized, Open-Label, Phase 1b/2 Study (LUC2001 JNJ-54767414). JTO Clin Res Rep 2, 100104 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Munoz P, Mittelbrunn M, de la Fuente H, Perez-Martinez M, Garcia-Perez A, Ariza-Veguillas A, Malavasi F, Zubiaur M, Sanchez-Madrid F, Sancho J, Antigen-induced clustering of surface CD38 and recruitment of intracellular CD38 to the immunologic synapse. Blood 111, 3653–3664 (2008). [DOI] [PubMed] [Google Scholar]
- 112.Partida-Sanchez S, Goodrich S, Kusser K, Oppenheimer N, Randall TD, Lund FE, Regulation of dendritic cell trafficking by the ADP-ribosyl cyclase CD38: impact on the development of humoral immunity. Immunity 20, 279–291 (2004). [DOI] [PubMed] [Google Scholar]
- 113.Stark R, Wesselink TH, Behr FM, Kragten NAM, Arens R, Koch-Nolte F, van Gisbergen K, van Lier RAW, T RM maintenance is regulated by tissue damage via P2RX7. Sci Immunol 3, (2018). [DOI] [PubMed] [Google Scholar]
- 114.Adriouch S, Hubert S, Pechberty S, Koch-Nolte F, Haag F, Seman M, NAD+ released during inflammation participates in T cell homeostasis by inducing ART2-mediated death of naive T cells in vivo. J Immunol 179, 186–194 (2007). [DOI] [PubMed] [Google Scholar]
- 115.Rissiek B, Haag F, Boyer O, Koch-Nolte F, Adriouch S, ADP-ribosylation of P2X7: a matter of life and death for regulatory T cells and natural killer T cells. Curr Top Microbiol Immunol 384, 107–126 (2015). [DOI] [PubMed] [Google Scholar]
- 116.Krebs C, Adriouch S, Braasch F, Koestner W, Leiter EH, Seman M, Lund FE, Oppenheimer N, Haag F, Koch-Nolte F, CD38 controls ADP-ribosyltransferase-2-catalyzed ADP-ribosylation of T cell surface proteins. J Immunol 174, 3298–3305 (2005). [DOI] [PubMed] [Google Scholar]
- 117.Haag F, Koch-Nolte F, Kuhl M, Lorenzen S, Thiele HG, Premature stop codons inactivate the RT6 genes of the human and chimpanzee species. J Mol Biol 243, 537–546 (1994). [DOI] [PubMed] [Google Scholar]
- 118.Wennerberg E, Mukherjee S, Spada S, Hung C, Agrusa CJ, Chen C, Valeta-Magara A, Rudqvist NP, Van Nest SJ, Kamel MK, Nasar A, Narula N, Mittal V, Markowitz GJ, Zhou XK, Adusumilli PS, Borczuk AC, White TE, Khan AG, Balderes PJ, Lorenz IC, Altorki N, Demaria S, McGraw TE, Stiles BM, Expression of the mono-ADP-ribosyltransferase ART1 by tumor cells mediates immune resistance in non-small cell lung cancer. Sci Transl Med 14, eabe8195 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Li J, Duran MA, Dhanota N, Chatila WK, Bettigole SE, Kwon J, Sriram RK, Humphries MP, Salto-Tellez M, James JA, Hanna MG, Melms JC, Vallabhaneni S, Litchfield K, Usaite I, Biswas D, Bareja R, Li HW, Martin ML, Dorsaint P, Cavallo JA, Li P, Pauli C, Gottesdiener L, DiPardo BJ, Hollmann TJ, Merghoub T, Wen HY, Reis-Filho JS, Riaz N, Su SM, Kalbasi A, Vasan N, Powell SN, Wolchok JD, Elemento O, Swanton C, Shoushtari AN, Parkes EE, Izar B, Bakhoum SF, Metastasis and Immune Evasion from Extracellular cGAMP Hydrolysis. Cancer Discov 11, 1212–1227 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Vanpouille-Box C, Demaria S, Formenti SC, Galluzzi L, Cytosolic DNA Sensing in Organismal Tumor Control. Cancer Cell 34, 361–378 (2018). [DOI] [PubMed] [Google Scholar]
- 121.Cordova AF, Ritchie C, Bohnert V, Li L, Human SLC46A2 Is the Dominant cGAMP Importer in Extracellular cGAMP-Sensing Macrophages and Monocytes. ACS Cent Sci 7, 1073–1088 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Lahey LJ, Mardjuki RE, Wen X, Hess GT, Ritchie C, Carozza JA, Bohnert V, Maduke M, Bassik MC, Li L, LRRC8A:C/E Heteromeric Channels Are Ubiquitous Transporters of cGAMP. Mol Cell 80, 578–591 e575 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Maltbaek JH, Cambier S, Snyder JM, Stetson DB, ABCC1 transporter exports the immunostimulatory cyclic dinucleotide cGAMP. Immunity 55, 1799–1812 e1794 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ablasser A, Schmid-Burgk JL, Hemmerling I, Horvath GL, Schmidt T, Latz E, Hornung V, Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature 503, 530–534 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Takahashi RU, Miyazaki H, Takeshita F, Yamamoto Y, Minoura K, Ono M, Kodaira M, Tamura K, Mori M, Ochiya T, Loss of microRNA-27b contributes to breast cancer stem cell generation by activating ENPP1. Nat Commun 6, 7318 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lau WM, Doucet M, Stadel R, Huang D, Weber KL, Kominsky SL, Enpp1: a potential facilitator of breast cancer bone metastasis. PLoS One 8, e66752 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Fontana GA, Gahlon HL, Mechanisms of replication and repair in mitochondrial DNA deletion formation. Nucleic Acids Res 48, 11244–11258 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Yamazaki T, Kirchmair A, Sato A, Buque A, Rybstein M, Petroni G, Bloy N, Finotello F, Stafford L, Navarro Manzano E, Ayala F. de la Pena E. Garcia-Martinez SC Formenti Z. Trajanoski Galluzzi, Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy. Nat Immunol 21, 1160–1171 (2020). [DOI] [PubMed] [Google Scholar]
- 129.Sun T, Li Y, Yang Y, Liu B, Cao Y, Yang W, Enhanced radiation-induced immunogenic cell death activates chimeric antigen receptor T cells by targeting CD39 against glioblastoma. Cell Death Dis 13, 875 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Loi S, Pommey S, Haibe-Kains B, Beavis PA, Darcy PK, Smyth MJ, Stagg J, CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc Natl Acad Sci U S A 110, 11091–11096 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Shanware NP, Trinh AT, Williams LM, Tibbetts RS, Coregulated ataxia telangiectasia-mutated and casein kinase sites modulate cAMP-response element-binding protein-coactivator interactions in response to DNA damage. J Biol Chem 282, 6283–6291 (2007). [DOI] [PubMed] [Google Scholar]
- 132.Bouquet F, Ousset M, Biard D, Fallone F, Dauvillier S, Frit P, Salles B, Muller C, A DNA-dependent stress response involving DNA-PK occurs in hypoxic cells and contributes to cellular adaptation to hypoxia. J Cell Sci 124, 1943–1951 (2011). [DOI] [PubMed] [Google Scholar]
- 133.Cam H, Easton JB, High A, Houghton PJ, mTORC1 signaling under hypoxic conditions is controlled by ATM-dependent phosphorylation of HIF-1alpha. Mol Cell 40, 509–520 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Fallone F, Britton S, Nieto L, Salles B, Muller C, ATR controls cellular adaptation to hypoxia through positive regulation of hypoxia-inducible factor 1 (HIF-1) expression. Oncogene 32, 4387–4396 (2013). [DOI] [PubMed] [Google Scholar]
- 135.Wu Y, Zheng J, Linden J, Holoshitz J, Genoprotective pathways. Part I. Extracellular signaling through G(s) protein-coupled adenosine receptors prevents oxidative DNA damage. Mutat Res 546, 93–102 (2004). [DOI] [PubMed] [Google Scholar]
- 136.Long JS, Crighton D, O’Prey J, Mackay G, Zheng L, Palmer TM, Gottlieb E, Ryan KM, Extracellular adenosine sensing-a metabolic cell death priming mechanism downstream of p53. Mol Cell 50, 394–406 (2013). [DOI] [PubMed] [Google Scholar]
- 137.Jacoberger-Foissac C, Cousineau I, Bareche Y, Allard D, Chrobak P, Allard B, Pommey S, Messaoudi N, McNicoll Y, Soucy G, Koseoglu S, Masia R, Lake AC, Seo H, Eeles CB, Rohatgi N, Robson SC, Turcotte S, Haibe-Kains B, Stagg J, CD73 Inhibits cGAS-STING and Cooperates with CD39 to Promote Pancreatic Cancer. Cancer Immunol Res 11, 56–71 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Kitabatake K, Kaji T, Tsukimoto M, Involvement of CD73 and A2B Receptor in Radiation-Induced DNA Damage Response and Cell Migration in Human Glioblastoma A172 Cells. Biol Pharm Bull 44, 197–210 (2021). [DOI] [PubMed] [Google Scholar]
- 139.Kitabatake K, Yoshida E, Kaji T, Tsukimoto M, Involvement of adenosine A2B receptor in radiation-induced translocation of epidermal growth factor receptor and DNA damage response leading to radioresistance in human lung cancer cells. Biochim Biophys Acta Gen Subj 1864, 129457 (2020). [DOI] [PubMed] [Google Scholar]
- 140.Searle JS, Schollaert KL, Wilkins BJ, Sanchez Y, The DNA damage checkpoint and PKA pathways converge on APC substrates and Cdc20 to regulate mitotic progression. Nat Cell Biol 6, 138–145 (2004). [DOI] [PubMed] [Google Scholar]
- 141.Soriano-Carot M, Quilis I, Bano MC, Igual JC, Protein kinase C controls activation of the DNA integrity checkpoint. Nucleic Acids Res 42, 7084–7095 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Bluwstein A, Kumar N, Leger K, Traenkle J, Oostrum J, Rehrauer H, Baudis M, Hottiger MO, PKC signaling prevents irradiation-induced apoptosis of primary human fibroblasts. Cell Death Dis 4, e498 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Mittal D, Sinha D, Barkauskas D, Young A, Kalimutho M, Stannard K, Caramia F, Haibe-Kains B, Stagg J, Khanna KK, Loi S, Smyth MJ, Adenosine 2B Receptor Expression on Cancer Cells Promotes Metastasis. Cancer Res 76, 4372–4382 (2016). [DOI] [PubMed] [Google Scholar]
- 144.Giacomelli C, Daniele S, Romei C, Tavanti L, Neri T, Piano I, Celi A, Martini C, Trincavelli ML, The A(2B) Adenosine Receptor Modulates the Epithelial- Mesenchymal Transition through the Balance of cAMP/PKA and MAPK/ERK Pathway Activation in Human Epithelial Lung Cells. Front Pharmacol 9, 54 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Volman Y, Hefetz R, Galun E, Rachmilewitz J, DNA damage alters EGFR signaling and reprograms cellular response via Mre-11. Sci Rep 12, 5760 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Estrada-Bernal A, Chatterjee M, Haque SJ, Yang L, Morgan MA, Kotian S, Morrell D, Chakravarti A, Williams TM, MEK inhibitor GSK1120212-mediated radiosensitization of pancreatic cancer cells involves inhibition of DNA double-strand break repair pathways. Cell Cycle 14, 3713–3724 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Liccardi G, Hartley JA, Hochhauser D, EGFR nuclear translocation modulates DNA repair following cisplatin and ionizing radiation treatment. Cancer Res 71, 1103–1114 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Lambert AW, Weinberg RA, Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat Rev Cancer 21, 325–338 (2021). [DOI] [PubMed] [Google Scholar]
- 149.Zhang P, Wei Y, Wang L, Debeb BG, Yuan Y, Zhang J, Yuan J, Wang M, Chen D, Sun Y, Woodward WA, Liu Y, Dean DC, Liang H, Hu Y, Ang KK, Hung MC, Chen J, Ma L, ATM-mediated stabilization of ZEB1 promotes DNA damage response and radioresistance through CHK1. Nat Cell Biol 16, 864–875 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Liu Q, Palomero L, Moore J, Guix I, Espin R, Aytes A, Mao JH, Paulovich AG, Whiteaker JR, Ivey RG, Iliakis G, Luo D, Chalmers AJ, Murnane J, Pujana MA, Barcellos-Hoff MH, Loss of TGFbeta signaling increases alternative end-joining DNA repair that sensitizes to genotoxic therapies across cancer types. Sci Transl Med 13, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Bareche Y, Pommey S, Carneiro M, Buisseret L, Cousineau I, Thebault P, Chrobak P, Communal L, Allard D, Robson SC, Mes-Masson AM, Provencher D, Lapointe R, Stagg J, High-dimensional analysis of the adenosine pathway in high-grade serous ovarian cancer. J Immunother Cancer 9, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Chalmin F, Mignot G, Bruchard M, Chevriaux A, Vegran F, Hichami A, Ladoire S, Derangere V, Vincent J, Masson D, Robson SC, Eberl G, Pallandre JR, Borg C, Ryffel B, Apetoh L, Rebe C, Ghiringhelli F, Stat3 and Gfi-1 transcription factors control Th17 cell immunosuppressive activity via the regulation of ectonucleotidase expression. Immunity 36, 362–373 (2012). [DOI] [PubMed] [Google Scholar]
- 153.Li Y, Liu Y, Chiang YJ, Huang F, Li Y, Li X, Ning Y, Zhang W, Deng H, Chen YG, DNA Damage Activates TGF-beta Signaling via ATM-c-Cbl-Mediated Stabilization of the Type II Receptor TbetaRII. Cell Rep 28, 735–745 e734 (2019). [DOI] [PubMed] [Google Scholar]
- 154.Liu Q, Ma L, Jones T, Palomero L, Pujana MA, Martinez-Ruiz H, Ha PK, Murnane J, Cuartas I, Seoane J, Baumann M, Linge A, Barcellos-Hoff MH, Subjugation of TGFbeta Signaling by Human Papilloma Virus in Head and Neck Squamous Cell Carcinoma Shifts DNA Repair from Homologous Recombination to Alternative End Joining. Clin Cancer Res 24, 6001–6014 (2018). [DOI] [PubMed] [Google Scholar]
- 155.Marcus A, Mao AJ, Lensink-Vasan M, Wang L, Vance RE, Raulet DH, Tumor-Derived cGAMP Triggers a STING-Mediated Interferon Response in Non-tumor Cells to Activate the NK Cell Response. Immunity 49, 754–763 e754 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Yoshida R, Saigi M, Tani T, Springer BF, Shibata H, Kitajima S, Mahadevan NR, Campisi M, Kim W, Kobayashi Y, Thai TC, Haratani K, Yamamoto Y, Sundararaman SK, Knelson EH, Vajdi A, Canadas I, Uppaluri R, Paweletz CP, Miret JJ, Lizotte PH, Gokhale PC, Janne PA, Barbie DA, MET-Induced CD73 Restrains STING-Mediated Immunogenicity of EGFR-Mutant Lung Cancer. Cancer Res 82, 4079–4092 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Allard D, Cousineau I, Ma EH, Allard B, Barèche Y, Fleury H, Stagg J, The CD73 immune checkpoint promotes tumor cell metabolic fitness. eLife 12, e84508 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Allard B, Allard D, Buisseret L, Stagg J, The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol 17, 611–629 (2020). [DOI] [PubMed] [Google Scholar]
- 159.Onyedibe KI, Wang M, Sintim HO, ENPP1, an Old Enzyme with New Functions, and Small Molecule Inhibitors-A STING in the Tale of ENPP1. Molecules 24, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Sidders B, Zhang P, Goodwin K, O’Connor G, Russell DL, Borodovsky A, Armenia J, McEwen R, Linghu B, Bendell JC, Bauer TM, Patel MR, Falchook GS, Merchant M, Pouliot G, Barrett JC, Dry JR, Woessner R, Sachsenmeier K, Adenosine Signaling Is Prognostic for Cancer Outcome and Has Predictive Utility for Immunotherapeutic Response. Clin Cancer Res 26, 2176–2187 (2020). [DOI] [PubMed] [Google Scholar]
- 161.Augustin RC, Leone RD, Naing A, Fong L, Bao R, Luke JJ, Next steps for clinical translation of adenosine pathway inhibition in cancer immunotherapy. J Immunother Cancer 10, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Herbst RS, Majem M, Barlesi F, Carcereny E, Chu Q, Monnet I, Sanchez-Hernandez A, Dakhil S, Camidge DR, Winzer L, Soo-Hoo Y, Cooper ZA, Kumar R, Bothos J, Aggarwal C, Martinez-Marti A, COAST: An Open-Label, Phase II, Multidrug Platform Study of Durvalumab Alone or in Combination With Oleclumab or Monalizumab in Patients With Unresectable, Stage III Non-Small-Cell Lung Cancer. J Clin Oncol, JCO2200227 (2022). [DOI] [PubMed]
- 163.Buisseret L, Loirat D, Aftimos PG, Punie K, Maurer C, Debien V, Goncalves A, Ghiringhelli F, Taylor D, Clatot F, van den Mooter TFA, Ferrero J, Bonnefoi H, Canon J, Duhoux FP, Bazan F, Kristanto P, de Azambuja E, Ignatiadis M, Piccart M, Primary endpoint results of SYNERGY, a randomized phase II trial, first-line chemo-immunotherapy trial of durvalumab, paclitaxel, and carboplatin with or without the anti-CD73 antibody oleclumab in patients with advanced or metastatic triple-negative breast cancer (TNBC). Ann Oncol 33, S808–S869 (2022). [Google Scholar]
- 164.Waickman AT, Alme A, Senaldi L, Zarek PE, Horton M, Powell JD, Enhancement of tumor immunotherapy by deletion of the A2A adenosine receptor. Cancer Immunol Immunother 61, 917–926 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Beavis PA, Milenkovski N, Henderson MA, John LB, Allard B, Loi S, Kershaw MH, Stagg J, Darcy PK, Adenosine Receptor 2A Blockade Increases the Efficacy of Anti-PD-1 through Enhanced Antitumor T-cell Responses. Cancer Immunol Res 3, 506–517 (2015). [DOI] [PubMed] [Google Scholar]
- 166.Cekic C, Sag D, Day YJ, Linden J, Extracellular adenosine regulates naive T cell development and peripheral maintenance. J Exp Med 210, 2693–2706 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Cekic C, Linden J, Adenosine A2A receptors intrinsically regulate CD8+ T cells in the tumor microenvironment. Cancer Res 74, 7239–7249 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Fong L, Hotson A, Powderly JD, Sznol M, Heist RS, Choueiri TK, George S, Hughes BGM, Hellmann MD, Shepard DR, Rini BI, Kummar S, Weise AM, Riese MJ, Markman B, Emens LA, Mahadevan D, Luke JJ, Laport G, Brody JD, Hernandez-Aya L, Bonomi P, Goldman JW, Berim L, Renouf DJ, Goodwin RA, Munneke B, Ho PY, Hsieh J, McCaffery I, Kwei L, Willingham SB, Miller RA, Adenosine 2A Receptor Blockade as an Immunotherapy for Treatment-Refractory Renal Cell Cancer. Cancer Discov 10, 40–53 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Cekic C, Sag D, Li Y, Theodorescu D, Strieter RM, Linden J, Adenosine A2B receptor blockade slows growth of bladder and breast tumors. J Immunol 188, 198–205 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Iannone R, Miele L, Maiolino P, Pinto A, Morello S, Blockade of A2b adenosine receptor reduces tumor growth and immune suppression mediated by myeloid-derived suppressor cells in a mouse model of melanoma. Neoplasia 15, 1400–1409 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]




